1
|
Ghahramani Almanghadim H, Karimi B, Valizadeh S, Ghaedi K. Biological functions and affected signaling pathways by Long Non-Coding RNAs in the immune system. Noncoding RNA Res 2025; 10:70-90. [PMID: 39315339 PMCID: PMC11417496 DOI: 10.1016/j.ncrna.2024.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 08/14/2024] [Accepted: 09/01/2024] [Indexed: 09/25/2024] Open
Abstract
Recently, the various regulative functions of long non-coding RNAs (LncRNAs) have been well determined. Recently, the vital role of LncRNAs as gene regulators has been identified in the immune system, especially in the inflammatory response. All cells of the immune system are governed by a complex and ever-changing gene expression program that is regulated through both transcriptional and post-transcriptional processes. LncRNAs regulate gene expression within the cell nucleus by influencing transcription or through post-transcriptional processes that affect the splicing, stability, or translation of messenger RNAs (mRNAs). Recent studies in immunology have revealed substantial alterations in the expression of lncRNAs during the activation of the innate immune system as well as the development, differentiation, and activation of T cells. These lncRNAs regulate key aspects of immune function, including the manufacturing of inflammatory molecules, cellular distinction, and cell movement. They do this by modulating protein-protein interactions or through base pairing with RNA and DNA. Here we review the current understanding of the mechanism of action of lncRNAs as novel immune-related regulators and their impact on physiological and pathological processes related to the immune system, including autoimmune diseases. We also highlight the emerging pattern of gene expression control in important research areas at the intersection between immunology and lncRNA biology.
Collapse
Affiliation(s)
| | - Bahareh Karimi
- Department of Cellular and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Sepehr Valizadeh
- Department of Internal Medicine, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Kamran Ghaedi
- Department of Cell and Molecular Biology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| |
Collapse
|
2
|
Jasim H, Ghafouri B, Ernberg M. The diurnal pattern of cytokines, chemokines and growth factors in human saliva-a pilot study. FRONTIERS IN DENTAL MEDICINE 2024; 5:1420081. [PMID: 39917641 PMCID: PMC11797753 DOI: 10.3389/fdmed.2024.1420081] [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: 05/07/2024] [Accepted: 10/31/2024] [Indexed: 02/09/2025] Open
Abstract
Background Understanding of possible periodicity of cytokines, chemokines and growth factors is of great interest and provide valuable information for research into pathophysiological mechanism of inflammatory disease and chronic pain. Significant efforts have been made to identify different analytes in saliva. For precision and accuracy in measurement and interpretation of results, it is crucial to know the source of variability, especially the circadian variation for the analytes. Objective The study aimed to analyze circadian variation in 71 inflammatory markers in both unstimulated and stimulated saliva, as well as plasma, from a sample of healthy individuals. Methods Ten young adults participated. Unstimulated and stimulated whole saliva were collected at 3-h intervals between between 7:30 am and 7:30 pm. Blood samples were drawn in connection with the first and last saliva collection. All samples were analyzed using the U-PLEX 71-Plex assay. Results The analysis showed distinct clustering of the 71 inflammatory mediators between plasma and saliva. Furthermore, differences were also observed between stimulated and unstimulated saliva. The proteins were clustered into three groups that expressed different circadian rhythms. These clusters were stable over time in stimulated saliva but showed significant variability in unstimulated saliva (P < 0.05). Conclusions These results suggest that time of the day could influence the detection and interpretation of inflammatory markers and collecting saliva samples at consistent times across participants will help control for the natural fluctuations in salivary composition. The results encourage further exploration of salivary diagnostics, particularly in understanding circadian rhythms and localized immune responses.
Collapse
Affiliation(s)
- Hajer Jasim
- Department of Orofacial Pain and Jaw Function, Public Dental Services, Folktandvården Stockholm, Eastmaninstitutet, Stockholm, Sweden
- Division of Oral Diagnostics and Rehabilitation, Department of Dental Medicine Karolinska Institutet, Huddinge, Sweden
| | - Bijar Ghafouri
- Department of Medical and Health Sciences, Pain and Rehabilitation Centre, Linköping University, Linköping, Sweden
| | - Malin Ernberg
- Division of Oral Diagnostics and Rehabilitation, Department of Dental Medicine Karolinska Institutet, Huddinge, Sweden
| |
Collapse
|
3
|
Vakili O, Adibi Sedeh P, Pourfarzam M. Metabolic biomarkers in irritable bowel syndrome diagnosis. Clin Chim Acta 2024; 560:119753. [PMID: 38821336 DOI: 10.1016/j.cca.2024.119753] [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: 04/14/2024] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/02/2024]
Abstract
Irritable bowel syndrome (IBS) is a chronic gastrointestinal (GI) disorder characterized by altered bowel habits and abdominal discomfort during defecation. It significantly impacts life quality and work productivity for those affected. Global data suggests a slightly higher prevalence in females than in males. Today, unambiguous diagnosis of IBS remains challenging due to the absence of a specific biochemical, histopathological, or radiological test. Current diagnosis relies heavily on thorough symptom evaluation. Efforts by the Rome committees have established standardized diagnostic criteria (Rome I-IV), improving consistency and clinical applicability. Recent studies in this framework, seem to have successfully employed metabolomics techniques to identify distinct metabolite profiles in breath and stool samples of IBS patients, differentiating them from healthy controls and those with other functional GI disorders, such as inflammatory bowel disease (IBD). Building on this success, researchers are investigating the presence of similar metabolites in easily accessible biofluids such as urine, potentially offering a less invasive diagnostic approach. Accordingly, this review focuses on key metabolites specifically detected in IBS patients' biological specimens, with a focus on urinary metabolites, using various methods, particularly mass spectrometry (MS)-based techniques, including gas chromatography-MS (GC-MS), liquid chromatography-tandem MS (LC-MS/MS), and capillary electrophoresis-MS (CE-MS) metabolomics assays. These findings may make provision for a new set of non-invasive biomarkers for IBS diagnosis and management.
Collapse
Affiliation(s)
- Omid Vakili
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Peyman Adibi Sedeh
- Gastroenterology and Hepatology Research Center, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Morteza Pourfarzam
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
4
|
Chiang S, Grogan T, Kamounah S, Wei F, Tayob N, Kim JY, Kyun Park J, Akin D, Elashoff DA, Pedersen AML, Song YW, Wong DTW, Chia D. Distinctive profile of monomeric and polymeric anti-SSA/Ro52 immunoglobulin A1 isoforms in saliva of patients with primary Sjögren's syndrome and Sicca. RMD Open 2024; 10:e003666. [PMID: 38599651 PMCID: PMC11015269 DOI: 10.1136/rmdopen-2023-003666] [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: 09/01/2023] [Accepted: 03/04/2024] [Indexed: 04/12/2024] Open
Abstract
OBJECTIVE Primary Sjögren's syndrome (pSS) is the second most common chronic autoimmune connective tissue disease. Autoantibodies, immunoglobulin (IgG) anti-SSA/Ro, in serum is a key diagnostic feature of pSS. Since pSS is a disease of the salivary gland, we investigated anti-SSA/Ro52 in saliva. METHODS Using a novel electrochemical detection platform, Electric Field-Induced Release and Measurement, we measured IgG/M/A, IgG, IgA, IgA isotypes (IgA1 and IgA2) and IgA1 subclasses (polymeric and monomeric IgA1) to anti-SSA/Ro52 in saliva supernatant of 34 pSS, 35 dry eyes and dry mouth (patients with Sicca) and 41 health controls. RESULTS Saliva IgG/M/A, IgG, IgA, IgA isotypes and IgA1 subclasses to anti-SSA/Ro52 differed significantly between pSS, non-pSS Sicca and healthy subjects. Elevated monomeric IgA1 was observed in patients with non-pSS Sicca while elevated polymeric IgA1 (pIgA1) was observed in patients with pSS. Salivary polymeric but not monomeric IgA1 (mIgA1) isoform correlated with focus score (r2=0.467, p=0.001) CONCLUSIONS: Salivary anti-Ro52 polymeric IgA1 isoform is associated with glandular inflammation in pSS, while salivary monomeric IgA1 is associated with Sicca. Whether IgA1 isotope switching plays a role in the progression of the Sicca to pSS warrants further investigation.
Collapse
Affiliation(s)
| | - Tristan Grogan
- Department of Medicine Statistics Core, University of California, Los Angeles, Los Angeles, California, USA
| | - Sarah Kamounah
- Oral Biology and Immunopathology/Oral Medicine & Pathology, University of Copenhagen Faculty of Health and Medical Sciences, Copenhagen N, Denmark
| | - Fang Wei
- UCLA School of Dentistry, Los Angeles, California, USA
| | - Nabihah Tayob
- Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Ju Yeon Kim
- Rheumatology, Seoul National University Hospital, Seoul, Korea
| | - Jin Kyun Park
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - David Akin
- UCLA School of Dentistry, Los Angeles, California, USA
| | - David A Elashoff
- Department of Medicine Statistics Core, University of California, Los Angeles, Los Angeles, California, USA
| | - Anne Marie Lynge Pedersen
- Section for Oral Biolology and Immunopathology, Oral Medicine and Pathology, Department of Odontology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Yeong Wook Song
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | | | - David Chia
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, California, USA
| |
Collapse
|
5
|
Guney Z, Altingoz SM, Has H, Serdar MA, Kurgan S. The impact of electronic cigarettes on peri-implant health: A systematic review and meta-analysis. J Dent 2024; 143:104883. [PMID: 38360396 DOI: 10.1016/j.jdent.2024.104883] [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: 12/12/2023] [Revised: 02/05/2024] [Accepted: 02/06/2024] [Indexed: 02/17/2024] Open
Abstract
OBJECTIVES Recent literature suggests that the use of electronic cigarette (e-cigarette) is a substantial contributing factor to the unsuccessful outcomes of dental implant procedures. Our aim was to systematically review the effect of e-cigarette use on clinical (PI, PD, BOP), radiographic (bone loss), and immunologic (IL-1β) peri‑implant parameters. DATA Main search terms used in combination: electronic cigarette, peri‑implantitis, vaping. SOURCES An electronic search was undertaken for MEDLINE, EMBASE, COCHRANE, and SCOPUS databases between 2017 and 2023. STUDY SELECTION The study protocol was developed according to PRISMA guidelines, and the focus question was formulated according to the PICO strategy. No restriction was accepted regarding language or year to avoid selection bias; the initial database search yielded 49 publications. Following the selection process, only seven studies met the inclusion criteria. Seven studies were statistically analyzed via MedCalc program. A pooled effect was deemed statistically significant if the p-value was less than 0.05. CONCLUSION Electronic cigarettes cause an increase in probing depth, bone loss, and the level of IL-1β, one of the bone destruction mediators in the tissues around the implant, and a decrease in bleeding on probing. CLINICAL SIGNIFICANCE E-cigarette is a potential risk factor for the healing process and the results of implant treatment, similar to cigarettes. Performing clinical research to evaluate the e-cigarette effect on peri‑implantitis in an age and gender-match population is needed.
Collapse
Affiliation(s)
- Zeliha Guney
- Department of Periodontology, Faculty of Dentistry, Ankara Medipol University, Ankara, Turkey
| | - S Merve Altingoz
- Department of Periodontology, Faculty of Dentistry, Lokman Hekim University, Ankara, Turkey
| | - Hande Has
- Graduate School of Health Sciences, Ankara University, Ankara, Turkey
| | - Muhittin A Serdar
- Department of Medical Biochemistry, School of Medicine, Acibadem University, İstanbul, Turkey
| | - Sivge Kurgan
- Department of Periodontology, Faculty of Dentistry, Ankara University, Ankara, Turkey.
| |
Collapse
|
6
|
Priya Aarthy A, Sen S, Srinivasan M, Muthukumar S, Madhanraj P, Akbarsha MA, Archunan G. Ectopic pregnancy: search for biomarker in salivary proteome. Sci Rep 2023; 13:16828. [PMID: 37803047 PMCID: PMC10558548 DOI: 10.1038/s41598-023-43791-7] [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: 04/21/2023] [Accepted: 09/28/2023] [Indexed: 10/08/2023] Open
Abstract
Ectopic pregnancy (EP) is associated with high maternal morbidity and mortality. Ultrasonography is the only dependable diagnostic tool for confirming an ectopic pregnancy. In view of inadequate early detection methods, women suffer from a high-life risk due to the severity of EP. Early detection of EP using pathological/molecular markers will possibly improve clinical diagnosis and patient management. Salivary proteins contain potential biomarkers for diagnosing and detecting various physiological and/or pathological conditions. Therefore, the present investigation was designed to explore the salivary proteome with special reference to EP. Gel-based protein separation was performed on saliva, followed by identification of proteins using Liquid Chromatography-Tandem Mass Spectrometry (LC-MS/MS). Totally, 326 proteins were identified in the salivary samples, among which 101 were found to be specific for ruptured ectopic pregnancy (EPR). Reactome analysis revealed innate immune system, neutrophil degranulation, cell surface interactions at the vascular wall, and FCERI-mediated NF-kB activation as the major pathways to which the salivary proteins identified during EPR are associated. Glutathione-S-transferase omega-1 (GSTO1) is specific for EPR and has been reported as a candidate biomarker in the serum of EPR patients. Therefore, saliva would be a potential source of diagnostic non-invasive protein biomarker(s) for EP. Intensive investigation on the salivary proteins specific to EP can potentially lead to setting up of a panel of candidate biomarkers and developing a non-invasive protein-based diagnostic kit.
Collapse
Affiliation(s)
- Archunan Priya Aarthy
- Department of Obstetrics and Gynecology, Rabindra Nath Tagore Medical College, Udaipur, Rajasthan, India.
- Department of Obstetrics and Gynecology, Saveetha Medical College and Hospital, Deemed University, Chennai, India.
| | - Sangeetha Sen
- Department of Obstetrics and Gynecology, Rabindra Nath Tagore Medical College, Udaipur, Rajasthan, India
| | - Mahalingam Srinivasan
- Department of Animal Science, Bharathidasan University, Tiruchirappalli, Tamil Nadu, India
| | - Subramanian Muthukumar
- Deparment of Biotechnology, School of Chemical & Biotechnology (SCBT), SASTRA Deemed University, Thanjavur, Tamil Nadu, India.
| | - Pakirisamy Madhanraj
- Department of Microbiology, Marudupandiyar College, Thanjavur, Tamil Nadu, India
| | - Mohammad Abdulkader Akbarsha
- Mahatma Gandhi-Doerenkamp Centre for Alternatives, Bharathidasan University, Tiruchchirappalli, India
- Department of Biotechnology & Microbiology, National College (Autonomous), Tiruchchirappalli, India
| | - Govindaraju Archunan
- Department of Animal Science, Bharathidasan University, Tiruchirappalli, Tamil Nadu, India.
- Marudupandiyar College, Thanjavur, Tamil Nadu, India.
| |
Collapse
|
7
|
Alt-Holland A, Huang X, Mendez T, Singh ML, Papas AS, Cimmino J, Bairos T, Tzavaras E, Foley E, Pagni SE, Baleja JD. Identification of Salivary Metabolic Signatures Associated with Primary Sjögren's Disease. Molecules 2023; 28:5891. [PMID: 37570863 PMCID: PMC10421170 DOI: 10.3390/molecules28155891] [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: 05/14/2023] [Revised: 07/26/2023] [Accepted: 07/31/2023] [Indexed: 08/13/2023] Open
Abstract
Sjögren's disease (SjD) is the second most prevalent autoimmune disorder that involves chronic inflammation of exocrine glands. Correct diagnosis of primary SjD (pSjD) can span over many years since disease symptoms manifest only in advanced stages of salivary and lachrymal glandular destruction, and consensus diagnostic methods have critical sensitivity and selectivity limitations. Using nuclear magnetic resonance (NMR) spectroscopy, we determined the composition of metabolites in unstimulated saliva samples from 30 pSjD subjects and 30 participants who do not have Sjögren's disease (non-Sjögren's control group, NS-C). Thirty-four metabolites were quantified in each sample, and analysis was conducted on both non-normalized (concentration) and normalized metabolomics data from all study participants (ages 23-78) and on an age-restricted subset of the data (ages 30-70) while applying false discovery rate correction in determining data significance. The normalized data of saliva samples from all study participants, and of the age-restricted subset, indicated significant increases in the levels of glucose, glycerol, taurine, and lactate, as well as significant decreases in the levels of 5-aminopentanoate, acetate, butyrate and propionate, in subjects with pSjD compared to subjects in the NS-C group. Additionally, a significant increase in choline was found only in the age-restricted subset, and a significant decrease in fucose was found only in the whole study population in normalized data of saliva samples from the pSjD group compared to the NS-C group. Metabolite concentration data of saliva samples from all study participants, but not from the age-restricted subset, indicated significant increases in the levels of glucose, glycerol, taurine, and lactate in subjects with pSjD compared to controls. The study showed that NMR metabolomics can be implemented in defining salivary metabolic signatures that are associated with disease status, and can contribute to differential analysis between subjects with pSjD and those who are not affected with this disease, in the clinic.
Collapse
Affiliation(s)
- Addy Alt-Holland
- Department of Endodontics, Tufts University School of Dental Medicine, One Kneeland Street, Boston, MA 02111, USA
- Tufts University School of Dental Medicine, One Kneeland Street, Boston, MA 02111, USA
| | - Xuejian Huang
- Program in Pharmacology and Drug Development, Tufts University Graduate School of Biomedical Sciences, 136 Harrison Avenue, Boston, MA 02111, USA
| | - Tatiana Mendez
- Tufts University School of Dental Medicine, One Kneeland Street, Boston, MA 02111, USA
| | - Mabi L. Singh
- Department of Diagnostics Sciences, Division of Oral Medicine, Tufts University School of Dental Medicine, One Kneeland Street, Boston, MA 02111, USA
| | - Athena S. Papas
- Department of Diagnostics Sciences, Division of Oral Medicine, Tufts University School of Dental Medicine, One Kneeland Street, Boston, MA 02111, USA
| | - Joseph Cimmino
- Department of Diagnostics Sciences, Division of Oral Medicine, Tufts University School of Dental Medicine, One Kneeland Street, Boston, MA 02111, USA
| | - Tiffany Bairos
- Department of Diagnostics Sciences, Division of Oral Medicine, Tufts University School of Dental Medicine, One Kneeland Street, Boston, MA 02111, USA
| | - Elizabeth Tzavaras
- Department of Diagnostics Sciences, Division of Oral Medicine, Tufts University School of Dental Medicine, One Kneeland Street, Boston, MA 02111, USA
| | - Elizabeth Foley
- Department of Diagnostics Sciences, Division of Oral Medicine, Tufts University School of Dental Medicine, One Kneeland Street, Boston, MA 02111, USA
| | - Sarah E. Pagni
- Department of Public Health and Community Service, Division of Biostatistics and Experimental Design, Tufts University School of Dental Medicine, One Kneeland Street, Boston, MA 02111, USA
| | - James D. Baleja
- Program in Pharmacology and Drug Development, Tufts University Graduate School of Biomedical Sciences, 136 Harrison Avenue, Boston, MA 02111, USA
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, USA
- Department of Medical Education, Tufts University Graduate School of Biomedical Sciences, 136 Harrison Avenue, Boston, MA 02111, USA
| |
Collapse
|
8
|
Kamounah S, Tayob N, Chiang S, Wei F, Park JK, Kwon HM, Feng Z, Chia D, Pedersen AML, Song YW, Wong DT. Immunoassay Detects Salivary Anti-SSA/Ro-52 Autoantibodies in Seronegative Patients with Primary Sjögren's Syndrome. Immunohorizons 2023; 7:554-561. [PMID: 37466644 PMCID: PMC10587502 DOI: 10.4049/immunohorizons.2300043] [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: 06/29/2023] [Accepted: 06/29/2023] [Indexed: 07/20/2023] Open
Abstract
The diagnostic work-up for Sjögren's syndrome is challenging and complex, including testing for serum autoantibodies to SSA/Ro and a labial salivary gland biopsy. Furthermore, the diagnosis is often delayed. In this study, we tested the hypothesis that anti-SSA/Ro autoantibodies are detectable in the saliva of patients with primary Sjögren's syndrome (pSS) because the disease affects the salivary glands, and these autoantibodies display greater discriminatory performance in saliva than in serum. SSA/Ro-52 Ags were used to develop what is, to our knowledge, a novel quantitative electrochemical-based immunoassay: the electric field-induced release and measurement (EFIRM) platform. The clinical utility was determined by measuring salivary anti-SSA/Ro-52 autoantibodies in patients with pSS and sicca (n = 34), patients without pSS with sicca (n = 35), and healthy subjects (n = 41). The statistical analysis of discrimination included the area under the receiver operating characteristic curve. Salivary anti-SSA/Ro-52 autoantibodies were measured in 94% (32 of 34) of patients with pSS with 85% (29 of 34) seropositivity. Four of the five seronegative patients with pSS had EFIRM-measurable anti-SSA/Ro-52 autoantibodies in saliva. Additionally, 60% (21 of 35) of the seronegative patients without pSS who had sicca had EFIRM-detectable SSA/Ro-52 autoantibodies in saliva, indicating the onset of autoimmune disease. Two of the 41 healthy control subjects had EFIRM-detectable SSA/Ro-52 autoantibodies in their saliva. Salivary SSA/Ro-52 autoantibodies significantly discriminated patients with pSS or patients with the initial stage of autoimmune disease from healthy subjects with an area under the receiver operating characteristic curve of 0.91. Our findings suggest that the proposed saliva SSA/Ro-52 immunoassay improves early and accurate diagnosis of seronegative patients with pSS and patients with early-onset autoimmune disease.
Collapse
Affiliation(s)
- Sarah Kamounah
- Center of Oral/Head & Neck Oncology Research, School of Dentistry, University of California, Los Angeles, Los Angeles, CA
- Department of Odontology, Section of Oral Biology and Immunopathology/Oral Medicine & Pathology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen N, Denmark
| | - Nabihah Tayob
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA
- Department of Medicine, Harvard Medical School, Boston, MA
| | - Samantha Chiang
- Center of Oral/Head & Neck Oncology Research, School of Dentistry, University of California, Los Angeles, Los Angeles, CA
| | - Fang Wei
- Center of Oral/Head & Neck Oncology Research, School of Dentistry, University of California, Los Angeles, Los Angeles, CA
| | - Jin Kyun Park
- Division of Rheumatology, Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea
| | - Hyun Mi Kwon
- Division of Rheumatology, Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea
| | - Ziding Feng
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - David Chia
- Department of Pathology & Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| | - Anne Marie Lynge Pedersen
- Department of Odontology, Section of Oral Biology and Immunopathology/Oral Medicine & Pathology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen N, Denmark
| | - Yeong Wook Song
- Division of Rheumatology, Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea
| | - David T.W. Wong
- Center of Oral/Head & Neck Oncology Research, School of Dentistry, University of California, Los Angeles, Los Angeles, CA
| |
Collapse
|
9
|
VURAL B, ULUDAĞ İ, İNCE B, ÖZYURT C, ÖZTÜRK F, SEZGİNTÜRK MK. Fluid-based wearable sensors: a turning point in personalized healthcare. Turk J Chem 2023; 47:944-967. [PMID: 38173754 PMCID: PMC10760819 DOI: 10.55730/1300-0527.3588] [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: 03/15/2023] [Revised: 10/31/2023] [Accepted: 05/22/2023] [Indexed: 01/05/2024] Open
Abstract
Nowadays, it has become very popular to develop wearable devices that can monitor biomarkers to analyze the health status of the human body more comprehensively and accurately. Wearable sensors, specially designed for home care services, show great promise with their ease of use, especially during pandemic periods. Scientists have conducted many innovative studies on new wearable sensors that can noninvasively and simultaneously monitor biochemical indicators in body fluids for disease prediction, diagnosis, and management. Using noninvasive electrochemical sensors, biomarkers can be detected in tears, saliva, perspiration, and skin interstitial fluid (ISF). In this review, biofluids used for noninvasive wearable sensor detection under four main headings, saliva, sweat, tears, and ISF-based wearable sensors, were examined in detail. This report analyzes nearly 50 recent articles from 2017 to 2023. Based on current research, this review also discusses the evolution of wearable sensors, potential implementation challenges, and future prospects.
Collapse
Affiliation(s)
- Berfin VURAL
- Department of Bioengineering, Faculty of Engineering, Çanakkale Onsekiz Mart University, Çanakkale,
Turkiye
| | - İnci ULUDAĞ
- Department of Bioengineering, Faculty of Engineering, Çanakkale Onsekiz Mart University, Çanakkale,
Turkiye
| | - Bahar İNCE
- Department of Bioengineering, Faculty of Engineering, Çanakkale Onsekiz Mart University, Çanakkale,
Turkiye
| | - Canan ÖZYURT
- Department of Chemistry and Chemical Processing Technologies, Lapseki Vocational School, Çanakkale Onsekiz Mart University, Çanakkale,
Turkiye
| | - Funda ÖZTÜRK
- Department of Chemistry, Faculty of Arts and Sciences, Tekirdağ Namık Kemal University, Tekirdağ,
Turkiye
| | - Mustafa Kemal SEZGİNTÜRK
- Department of Bioengineering, Faculty of Engineering, Çanakkale Onsekiz Mart University, Çanakkale,
Turkiye
| |
Collapse
|
10
|
Kamounah S, Sembler-Møller ML, Nielsen CH, Pedersen AML. Sjögren's syndrome: novel insights from proteomics and miRNA expression analysis. Front Immunol 2023; 14:1183195. [PMID: 37275849 PMCID: PMC10232878 DOI: 10.3389/fimmu.2023.1183195] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 05/09/2023] [Indexed: 06/07/2023] Open
Abstract
Introduction Sjögren's syndrome (SS) is a systemic autoimmune disease, which affects the exocrine glands leading to glandular dysfunction and, particularly, symptoms of oral and ocular dryness. The aetiology of SS remains unclear, and the disease lacks distinctive clinical features. The current diagnostic work-up is complex, invasive and often time-consuming. Thus, there is an emerging need for identifying disease-specific and, ideally, non-invasive immunological and molecular biomarkers that can simplify the diagnostic process, allow stratification of patients, and assist in monitoring the disease course and outcome of therapeutic intervention in SS. Methods This systematic review addresses the use of proteomics and miRNA-expression profile analyses in this regard. Results and discussion Out of 272 papers that were identified and 108 reviewed, a total of 42 papers on proteomics and 23 papers on miRNA analyses in saliva, blood and salivary gland tissue were included in this review. Overall, the proteomic and miRNA studies revealed considerable variations with regard to candidate biomarker proteins and miRNAs, most likely due to variation in sample size, processing and analytical methods, but also reflecting the complexity of SS and patient heterogeneity. However, interesting novel knowledge has emerged and further validation is needed to confirm their potential role as biomarkers in SS.
Collapse
Affiliation(s)
- Sarah Kamounah
- Section for Oral Biology and Immunopathology/Oral Medicine, Department of Odontology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Maria Lynn Sembler-Møller
- Section for Oral Biology and Immunopathology/Oral Medicine, Department of Odontology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Claus Henrik Nielsen
- Section for Oral Biology and Immunopathology/Oral Medicine, Department of Odontology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Center for Rheumatology and Spine Diseases, University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Anne Marie Lynge Pedersen
- Section for Oral Biology and Immunopathology/Oral Medicine, Department of Odontology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
11
|
Liu A, Hefley B, Escandon P, Nicholas SE, Karamichos D. Salivary Exosomes in Health and Disease: Future Prospects in the Eye. Int J Mol Sci 2023; 24:ijms24076363. [PMID: 37047335 PMCID: PMC10094317 DOI: 10.3390/ijms24076363] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/21/2023] [Accepted: 03/24/2023] [Indexed: 03/30/2023] Open
Abstract
Exosomes are a group of vesicles that package and transport DNA, RNA, proteins, and lipids to recipient cells. They can be derived from blood, saliva, urine, and/or other biological tissues. Their impact on several diseases, such as neurodegenerative, autoimmune, and ocular diseases, have been reported, but not fully unraveled. The exosomes that are derived from saliva are less studied, but offer significant advantages over exosomes from other sources, due to their accessibility and ease of collection. Thus, their role in the pathophysiology of diseases is largely unknown. In the context of ocular diseases, salivary exosomes have been under-utilized, thus creating an enormous gap in the literature. The current review discusses the state of exosomes research on systemic and ocular diseases and highlights the role and potential of salivary exosomes as future ocular therapeutic vehicles.
Collapse
Affiliation(s)
- Angela Liu
- Texas College of Osteopathic Medicine, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX 76107, USA
- North Texas Eye Research Institute, University of North Texas Health Science Center, 3430 Camp Bowie Blvd., Fort Worth, TX 76107, USA
| | - Brenna Hefley
- North Texas Eye Research Institute, University of North Texas Health Science Center, 3430 Camp Bowie Blvd., Fort Worth, TX 76107, USA
- Department of Pharmaceutical Sciences, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX 76107, USA
| | - Paulina Escandon
- North Texas Eye Research Institute, University of North Texas Health Science Center, 3430 Camp Bowie Blvd., Fort Worth, TX 76107, USA
- Department of Pharmaceutical Sciences, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX 76107, USA
| | - Sarah E. Nicholas
- North Texas Eye Research Institute, University of North Texas Health Science Center, 3430 Camp Bowie Blvd., Fort Worth, TX 76107, USA
- Department of Pharmaceutical Sciences, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX 76107, USA
| | - Dimitrios Karamichos
- North Texas Eye Research Institute, University of North Texas Health Science Center, 3430 Camp Bowie Blvd., Fort Worth, TX 76107, USA
- Department of Pharmaceutical Sciences, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX 76107, USA
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX 76107, USA
- Correspondence: ; Tel.: +1-817-735-2101
| |
Collapse
|
12
|
Liu Y, Zhu J, Ding L. Involvement of RNA methylation modification patterns mediated by m7G, m6A, m5C and m1A regulators in immune microenvironment regulation of Sjögren's syndrome. Cell Signal 2023; 106:110650. [PMID: 36935085 DOI: 10.1016/j.cellsig.2023.110650] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 03/03/2023] [Indexed: 03/18/2023]
Abstract
Keratoconjunctivitis is the most common complication of Sjögren's syndrome (SS). It has always been a hot research topic due to its complex pathogenesis. A further understanding of keratoconjunctiva xerosis can be obtained by studying the primary diseases. 7-Methylguanine (m7G), N6-methyladenosine (m6A), 5-methylcytosine (m5C), and N1-methyladenosine (m1A) are newly discovered epigenetic mechanisms involved in the development of SS. This study aimed to investigate the effects of m7G, m6A, m5C, and m1A modifications on the immune microenvironment of SS. Three microarray datasets were downloaded from the Gene Omnibus Expression (GEO) database, including 56 SS samples and 35 normal samples. Then, genes with m7G, m6A, m5C, and m1A methylation were explored, and the RNA modification patterns mediated by 59 m7G, m6A, m5C, and m1A regulators were summarized. The effects of m7G, m6A, m5C, and m1A modifications on immune infiltrating cells were discussed. Eukaryotic translation initiation factor 3 subunit D(EIF3D) was closely related to monocytes, and the expression of EIF3D was higher in SS with less monocytes. Two distinct patterns of RNA modification mediated by the 59 m7G, m6A, m5C, and m1A regulators were also identified, which infiltrated immune cells differently. Moreover, the two distinct RNA patterns were enriched in different signaling pathways, and their biological functions were explored. The findings revealed that m7G, m6A, m5C, and m1A modifications played vital roles in the diversity and complexity of the immune microenvironment in SS.
Collapse
Affiliation(s)
- Yuxiu Liu
- Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region, China.
| | - Jianing Zhu
- Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Lin Ding
- Xinjiang Uygur Autonomous Region People's Hospital, 91 Longquan Street, Urumqi, Xinjiang Uygur Autonomous Region, China.
| |
Collapse
|
13
|
Katsiougiannis S, Stergiopoulos A, Moustaka K, Havaki S, Samiotaki M, Stamatakis G, Tenta R, Skopouli FN. Salivary gland epithelial cell in Sjögren's syndrome: Metabolic shift and altered mitochondrial morphology toward an innate immune cell function. J Autoimmun 2023; 136:103014. [PMID: 36898185 DOI: 10.1016/j.jaut.2023.103014] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 02/19/2023] [Indexed: 03/10/2023]
Abstract
Salivary gland epithelial cells (SGEC) are the main targets of the autoimmune reactivity in Sjögren's syndrome (SS). This study aimed to investigate the core proteomic differences between SS and Control- (Ct) -derived SGEC. Proteome analysis of cultured SGEC from five SS patients and four Ct was performed in a label-free quantitation format (LFQ). Electron microscopy was applied for analysis of the mitochondrial ultrastructure of SGEC in minor salivary gland sections from six SS patients and four Ct. Four hundred seventy-four proteins were identified differentially abundant in SS- compared to Ct-SGEC. After proteomic analysis, two distinct protein expression patterns were revealed. Gene ontology (GO) pathway analysis of each protein block revealed that the cluster with highly abundant proteins in SS-SGEC showed enrichment in pathways associated with membrane trafficking, exosome-mediated transport and exocytosis as well as innate immunity related mainly to neutrophil degranulation. In contrast, the low abundance protein cluster in SS-SGEC was enriched for proteins regulating the translational process of proteins related to metabolic pathways associated to mitochondria. Electron microscopy showed decreased total number of mitochondria in SS-SGEC, which appeared elongated and swollen with less and abnormal cristae compared to Ct-SGEC mitochondria. This study defines, for the first time, the core proteomic differences of SGEC between SS and Ct, substantiates the metamorphosis of SGEC into an innate immune cell and reveals that these cells are translationally shifted towards metabolism rewiring. These metabolic alterations are related mainly to mitochondria and are mirrored in situ with heavy morphological changes.
Collapse
Affiliation(s)
- S Katsiougiannis
- Department of Nutrition and Dietetics, School of Health Sciences & Education, Harokopio University, Athens, Greece; Laboratory of Autoimmunity, Biomedical Research Foundation of the Academy of Athens, Greece
| | - A Stergiopoulos
- Department of Nutrition and Dietetics, School of Health Sciences & Education, Harokopio University, Athens, Greece
| | - K Moustaka
- Department of Nutrition and Dietetics, School of Health Sciences & Education, Harokopio University, Athens, Greece
| | - S Havaki
- Department of Histology and Embryology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - M Samiotaki
- Institute for Bio-Innovation, Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece
| | - G Stamatakis
- Institute for Bio-Innovation, Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece
| | - R Tenta
- Department of Nutrition and Dietetics, School of Health Sciences & Education, Harokopio University, Athens, Greece
| | - F N Skopouli
- Department of Nutrition and Dietetics, School of Health Sciences & Education, Harokopio University, Athens, Greece; Euroclinic of Athens, Athens, Greece.
| |
Collapse
|
14
|
Metabolic Profile of Whole Unstimulated Saliva in Patients with Sjögren’s Syndrome. Metabolites 2023; 13:metabo13030348. [PMID: 36984788 PMCID: PMC10054882 DOI: 10.3390/metabo13030348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 02/22/2023] [Accepted: 02/24/2023] [Indexed: 03/03/2023] Open
Abstract
(1) Background: Primary Sjögren’s Syndrome (pSS) is a multi-system autoimmune disease that involves the exocrine glands. Lymphocytes infiltrate the gland tissue, leading to anatomical modification and hypofunction. Even if the prognosis of pSS is favorable, quality of life is typically reduced due to the diverse manifestations of the disease. The aim of this study is to compare the salivary metabolomes of pSS with healthy controls (HCs). (2) Methods: Seven cases were selected from a cohort of pSS patients, and six age- and sex-matched HCs were recruited from a cohort of volunteers. Whole unstimulated saliva was collected for NMR analysis. Our metabolomic analysis focused on 360 ms total echo 1D 1H NMR CPMG spectra. Metabolites detected with CPMG NMR spectra were assigned through 2D NMR spectra (COSY, TOCSY, and HSQC). (3) Results: About 50 metabolites were detected and assigned. Unsupervised exploratory PCA returned partial clustering, and PLS-DA improved the separation between pSS and HCs, highlighting a pool of metabolites distinctly describing each group. (4) Conclusions: Despite the limited number of samples, the presented preliminary data are promising. PLS-DA indicated well-defined group separation, suggesting that the application of 1H-NMR metabolomics is suitable for the study of pSS.
Collapse
|
15
|
Mangolini V, Gualerzi A, Picciolini S, Rodà F, Del Prete A, Forleo L, Rossetto RA, Bedoni M. Biochemical Characterization of Human Salivary Extracellular Vesicles as a Valuable Source of Biomarkers. BIOLOGY 2023; 12:227. [PMID: 36829504 PMCID: PMC9953587 DOI: 10.3390/biology12020227] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 01/27/2023] [Accepted: 01/30/2023] [Indexed: 02/04/2023]
Abstract
Extracellular vesicles (EVs) are natural nanoparticles secreted under physiological and pathological conditions. Thanks to their diagnostic potential, EVs are increasingly being studied as biomarkers of a variety of diseases, including neurological disorders. To date, most studies on EV biomarkers use blood as the source, despite different disadvantages that may cause an impure isolation of the EVs. In the present article, we propose the use of saliva as a valuable source of EVs that could be studied as biomarkers in an easily accessible biofluid. Using a comparable protocol for the isolation of EVs from both liquid biopsies, salivary EVs showed greater purity in terms of co-isolates (evaluated by nanoparticle tracking analysis and Conan test). In addition, Raman spectroscopy was used for the identification of the overall biochemical composition of EVs coming from the two different biofluids. Even considering the limited amount of EVs that can be isolated from saliva, the use of Raman spectroscopy was not hampered, and it was able to provide a comprehensive characterization of EVs in a high throughput and repeatable manner. Raman spectroscopy can thus represent a turning point in the application of salivary EVs in clinics, taking advantage of the simple method of collection of the liquid biopsy and of the quick, sensitive and label-free biophotonics-based approach.
Collapse
Affiliation(s)
- Valentina Mangolini
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, 20148 Milano, Italy
- Dipartimento di Medicina Molecolare e Traslazionale, Università degli Studi di Brescia, 25122 Brescia, Italy
| | - Alice Gualerzi
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, 20148 Milano, Italy
| | | | - Francesca Rodà
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, 20148 Milano, Italy
- Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, 42100 Modena, Italy
| | | | - Luana Forleo
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, 20148 Milano, Italy
| | | | - Marzia Bedoni
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, 20148 Milano, Italy
| |
Collapse
|
16
|
Applications of Mass Spectrometry in Dentistry. Biomedicines 2023; 11:biomedicines11020286. [PMID: 36830822 PMCID: PMC9953492 DOI: 10.3390/biomedicines11020286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/16/2023] [Accepted: 01/18/2023] [Indexed: 01/21/2023] Open
Abstract
Mass Spectrometry (MS) is one of the fastest-developing methods in analytical instrumentation. As a highly sensitive, universal detector, it can identify known and unknown compounds, which can indeed be found in a minimal concentration. This review aims to highlight the significant milestones in MS applications in dentistry during recent decades. MS can be applied in three different fields of dentistry: (1) in research of dental materials and chemical agents, (2) in laboratory analysis of biospecimens, and (3) as a real-time diagnostic tool in service of oral surgery and pathology. MS applications on materials and agents may focus on numerous aspects, such as their clinical behavior, possible toxicity, or antimicrobial properties. MS is also a valuable, non-invasive tool for biomarkers' detection in saliva and has found great application in -omics technologies as it achieves efficient structure-finding in metabolites. As metabolites are located beyond the central dogma, this technique can provide a complete understanding of cellular functions. Thus, it is possible to determine the biological profile in normal and pathological conditions, detect various oral or systematic diseases and conditions, and predict their course. Lastly, some promising advances concerning the surgical approach to potentially oral malignant or malignant disorders exist. This breakthrough method provides a comprehensive approach to dental materials research and biomarker discovery in dental and craniofacial tissues. The current availability of various 'OMIC' approaches paves the way for individualized dentistry and provides suggestions for clinical applications in the point-of-care hubs.
Collapse
|
17
|
Shang YF, Shen YY, Zhang MC, Lv MC, Wang TY, Chen XQ, Lin J. Progress in salivary glands: Endocrine glands with immune functions. Front Endocrinol (Lausanne) 2023; 14:1061235. [PMID: 36817607 PMCID: PMC9935576 DOI: 10.3389/fendo.2023.1061235] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 01/09/2023] [Indexed: 02/05/2023] Open
Abstract
The production and secretion of saliva is an essential function of the salivary glands. Saliva is a complicated liquid with different functions, including moistening, digestion, mineralization, lubrication, and mucosal protection. This review focuses on the mechanism and neural regulation of salivary secretion, and saliva is secreted in response to various stimuli, including odor, taste, vision, and mastication. The chemical and physical properties of saliva change dynamically during physiological and pathophysiological processes. Moreover, the central nervous system modulates salivary secretion and function via various neurotransmitters and neuroreceptors. Smell, vision, and taste have been investigated for the connection between salivation and brain function. The immune and endocrine functions of the salivary glands have been explored recently. Salivary glands play an essential role in innate and adaptive immunity and protection. Various immune cells such as B cells, T cells, macrophages, and dendritic cells, as well as immunoglobins like IgA and IgG have been found in salivary glands. Evidence supports the synthesis of corticosterone, testosterone, and melatonin in salivary glands. Saliva contains many potential biomarkers derived from epithelial cells, gingival crevicular fluid, and serum. High level of matrix metalloproteinases and cytokines are potential markers for oral carcinoma, infectious disease in the oral cavity, and systemic disease. Further research is required to monitor and predict potential salivary biomarkers for health and disease in clinical practice and precision medicine.
Collapse
Affiliation(s)
- Yu Feng Shang
- Department of Stomatology, Key Laboratory of Oral Biomedical Research of Zhejiang Province, The First Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang University School of Stomatology, Hangzhou, China
| | - Yi Yang Shen
- Department of Stomatology, Key Laboratory of Oral Biomedical Research of Zhejiang Province, The First Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang University School of Stomatology, Hangzhou, China
| | - Meng Chen Zhang
- National Health Commission and Chinese Academy of Medical Sciences Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Min Chao Lv
- Department of Orthopedics, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital, Quzhou, China
| | - Tong Ying Wang
- National Health Commission and Chinese Academy of Medical Sciences Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
- Department of Neurobiology, Department of Neurology of the Second Affiliated Hospital, School of Brain Science and Brain Medicine, Hangzhou, China
| | - Xue Qun Chen
- National Health Commission and Chinese Academy of Medical Sciences Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
- Department of Neurobiology, Department of Neurology of the Second Affiliated Hospital, School of Brain Science and Brain Medicine, Hangzhou, China
| | - Jun Lin
- Department of Stomatology, Key Laboratory of Oral Biomedical Research of Zhejiang Province, The First Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang University School of Stomatology, Hangzhou, China
- *Correspondence: Jun Lin,
| |
Collapse
|
18
|
Khatri B, Tessneer KL, Rasmussen A, Aghakhanian F, Reksten TR, Adler A, Alevizos I, Anaya JM, Aqrawi LA, Baecklund E, Brun JG, Bucher SM, Eloranta ML, Engelke F, Forsblad-d’Elia H, Glenn SB, Hammenfors D, Imgenberg-Kreuz J, Jensen JL, Johnsen SJA, Jonsson MV, Kvarnström M, Kelly JA, Li H, Mandl T, Martín J, Nocturne G, Norheim KB, Palm Ø, Skarstein K, Stolarczyk AM, Taylor KE, Teruel M, Theander E, Venuturupalli S, Wallace DJ, Grundahl KM, Hefner KS, Radfar L, Lewis DM, Stone DU, Kaufman CE, Brennan MT, Guthridge JM, James JA, Scofield RH, Gaffney PM, Criswell LA, Jonsson R, Eriksson P, Bowman SJ, Omdal R, Rönnblom L, Warner B, Rischmueller M, Witte T, Farris AD, Mariette X, Alarcon-Riquelme ME, Shiboski CH, Wahren-Herlenius M, Ng WF, Sivils KL, Adrianto I, Nordmark G, Lessard CJ. Genome-wide association study identifies Sjögren's risk loci with functional implications in immune and glandular cells. Nat Commun 2022; 13:4287. [PMID: 35896530 PMCID: PMC9329286 DOI: 10.1038/s41467-022-30773-y] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 05/17/2022] [Indexed: 02/06/2023] Open
Abstract
Sjögren's disease is a complex autoimmune disease with twelve established susceptibility loci. This genome-wide association study (GWAS) identifies ten novel genome-wide significant (GWS) regions in Sjögren's cases of European ancestry: CD247, NAB1, PTTG1-MIR146A, PRDM1-ATG5, TNFAIP3, XKR6, MAPT-CRHR1, RPTOR-CHMP6-BAIAP6, TYK2, SYNGR1. Polygenic risk scores yield predictability (AUROC = 0.71) and relative risk of 12.08. Interrogation of bioinformatics databases refine the associations, define local regulatory networks of GWS SNPs from the 95% credible set, and expand the implicated gene list to >40. Many GWS SNPs are eQTLs for genes within topologically associated domains in immune cells and/or eQTLs in the main target tissue, salivary glands.
Collapse
Affiliation(s)
- Bhuwan Khatri
- grid.274264.10000 0000 8527 6890Genes and Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK USA
| | - Kandice L. Tessneer
- grid.274264.10000 0000 8527 6890Genes and Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK USA
| | - Astrid Rasmussen
- grid.274264.10000 0000 8527 6890Genes and Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK USA
| | - Farhang Aghakhanian
- grid.274264.10000 0000 8527 6890Genes and Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK USA
| | - Tove Ragna Reksten
- grid.274264.10000 0000 8527 6890Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK USA ,grid.7914.b0000 0004 1936 7443Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Adam Adler
- grid.274264.10000 0000 8527 6890NGS Core Laboratory, Oklahoma Medical Research Foundation, Oklahoma City, OK USA
| | - Ilias Alevizos
- grid.419633.a0000 0001 2205 0568Salivary Disorder Unit, National Institute of Dental and Craniofacial Research, Bethesda, MD USA
| | - Juan-Manuel Anaya
- grid.412191.e0000 0001 2205 5940Center for Autoimmune Diseases Research (CREA), Universidad del Rosario, Bogotá, Colombia
| | - Lara A. Aqrawi
- grid.5510.10000 0004 1936 8921Department of Oral Surgery and Oral Medicine, Faculty of Dentistry, University of Oslo, Oslo, Norway ,grid.457625.70000 0004 0383 3497Department of Health Sciences, Kristiania University College, Oslo, Norway
| | - Eva Baecklund
- grid.8993.b0000 0004 1936 9457Department of Medical Sciences, Rheumatology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Johan G. Brun
- grid.7914.b0000 0004 1936 7443Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Sara Magnusson Bucher
- grid.15895.300000 0001 0738 8966Department of Rheumatology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Maija-Leena Eloranta
- grid.8993.b0000 0004 1936 9457Department of Medical Sciences, Rheumatology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Fiona Engelke
- grid.10423.340000 0000 9529 9877Department of Rheumatology and Immunology, Hannover Medical School, Hannover, Germany
| | - Helena Forsblad-d’Elia
- grid.8761.80000 0000 9919 9582Department of Rheumatology and Inflammation Research, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Stuart B. Glenn
- grid.274264.10000 0000 8527 6890Genes and Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK USA
| | - Daniel Hammenfors
- grid.412008.f0000 0000 9753 1393Department of Rheumatology, Haukeland University Hospital, Bergen, Norway
| | - Juliana Imgenberg-Kreuz
- grid.8993.b0000 0004 1936 9457Department of Medical Sciences, Rheumatology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Janicke Liaaen Jensen
- grid.5510.10000 0004 1936 8921Department of Oral Surgery and Oral Medicine, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - Svein Joar Auglænd Johnsen
- grid.412835.90000 0004 0627 2891Department of Internal Medicine, Clinical Immunology Unit, Stavanger University Hospital, Stavanger, Norway
| | - Malin V. Jonsson
- grid.7914.b0000 0004 1936 7443Department of Clinical Science, University of Bergen, Bergen, Norway ,grid.7914.b0000 0004 1936 7443Section for Oral and Maxillofacial Radiology, Department of Clinical Dentistry, Medical Faculty, University of Bergen, Bergen, Norway
| | - Marika Kvarnström
- grid.4714.60000 0004 1937 0626Rheumatology Unity, Department of Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden ,grid.425979.40000 0001 2326 2191Academic Specialist Center, Center for Rheumatology and Studieenheten, Stockholm Health Services, Region Stockholm, Sweden
| | - Jennifer A. Kelly
- grid.274264.10000 0000 8527 6890Genes and Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK USA
| | - He Li
- grid.274264.10000 0000 8527 6890Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK USA ,grid.505430.7Translational Sciences, The Janssen Pharmaceutical Companies of Johnson & Johnson, Spring House, PA USA
| | - Thomas Mandl
- grid.4514.40000 0001 0930 2361Rheumatology, Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Javier Martín
- grid.4711.30000 0001 2183 4846Instituto de Biomedicina y Parasitología López-Neyra, Consejo Superior de Investigaciones Científicas (CSIC), Granada, Spain
| | - Gaétane Nocturne
- grid.413784.d0000 0001 2181 7253Université Paris-Saclay, Assistance Publique–Hôpitaux de Paris (AP-HP), Hôpital Bicêtre, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1184, Le Kremlin Bicêtre, France
| | - Katrine Brække Norheim
- grid.7914.b0000 0004 1936 7443Department of Clinical Science, University of Bergen, Bergen, Norway ,grid.412835.90000 0004 0627 2891Department of Rheumatology, Stavanger University Hospital, Stavanger, Norway
| | - Øyvind Palm
- grid.5510.10000 0004 1936 8921Department of Rheumatology, University of Oslo, Oslo, Norway
| | - Kathrine Skarstein
- grid.7914.b0000 0004 1936 7443Department of Clinical Science, University of Bergen, Bergen, Norway ,grid.412008.f0000 0000 9753 1393Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Anna M. Stolarczyk
- grid.274264.10000 0000 8527 6890Genes and Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK USA
| | - Kimberly E. Taylor
- grid.266102.10000 0001 2297 6811Department of Medicine, Russell/Engleman Rheumatology Research Center, University of California San Francisco, San Francisco, California USA
| | - Maria Teruel
- grid.4489.10000000121678994Genyo, Center for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, Granada, Spain
| | - Elke Theander
- grid.411843.b0000 0004 0623 9987Department of Rheumatology, Skåne University Hospital, Malmö, Sweden ,Medical Affairs, Jannsen-Cilag EMEA (Europe/Middle East/Africa), Beerse, Belgium
| | - Swamy Venuturupalli
- grid.50956.3f0000 0001 2152 9905Division of Rheumatology, Cedars-Sinai Medical Center, Los Angeles, CA USA ,grid.19006.3e0000 0000 9632 6718David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA USA
| | - Daniel J. Wallace
- grid.50956.3f0000 0001 2152 9905Division of Rheumatology, Cedars-Sinai Medical Center, Los Angeles, CA USA ,grid.19006.3e0000 0000 9632 6718David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA USA
| | - Kiely M. Grundahl
- grid.274264.10000 0000 8527 6890Genes and Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK USA
| | | | - Lida Radfar
- grid.266900.b0000 0004 0447 0018Oral Diagnosis and Radiology Department, University of Oklahoma College of Dentistry, Oklahoma City, OK USA
| | - David M. Lewis
- grid.266900.b0000 0004 0447 0018Department of Oral and Maxillofacial Pathology, University of Oklahoma College of Dentistry, Oklahoma City, OK USA
| | - Donald U. Stone
- grid.266902.90000 0001 2179 3618Department of Ophthalmology, Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
| | - C. Erick Kaufman
- grid.266902.90000 0001 2179 3618Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
| | - Michael T. Brennan
- grid.239494.10000 0000 9553 6721Department of Oral Medicine/Oral & Maxillofacial Surgery, Atrium Health Carolinas Medical Center, Charlotte, NC USA ,grid.241167.70000 0001 2185 3318Department of Otolaryngology/Head and Neck Surgery, Wake Forest University School of Medicine, Winston-Salem, NC USA
| | - Joel M. Guthridge
- grid.274264.10000 0000 8527 6890Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK USA ,grid.266902.90000 0001 2179 3618Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
| | - Judith A. James
- grid.274264.10000 0000 8527 6890Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK USA ,grid.266902.90000 0001 2179 3618Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
| | - R. Hal Scofield
- grid.274264.10000 0000 8527 6890Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK USA ,grid.266902.90000 0001 2179 3618Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA ,grid.413864.c0000 0004 0420 2582US Department of Veterans Affairs Medical Center, Oklahoma City, OK USA
| | - Patrick M. Gaffney
- grid.274264.10000 0000 8527 6890Genes and Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK USA
| | - Lindsey A. Criswell
- grid.266102.10000 0001 2297 6811Department of Medicine, Russell/Engleman Rheumatology Research Center, University of California San Francisco, San Francisco, California USA ,grid.266102.10000 0001 2297 6811Institute of Human Genetics (IHG), University of California San Francisco, San Francisco, CA USA ,grid.280128.10000 0001 2233 9230Genomics of Autoimmune Rheumatic Disease Section, National Human Genome Research Institute, NIH, Bethesda, MD USA
| | - Roland Jonsson
- grid.7914.b0000 0004 1936 7443Department of Clinical Science, University of Bergen, Bergen, Norway ,grid.412008.f0000 0000 9753 1393Department of Rheumatology, Haukeland University Hospital, Bergen, Norway
| | - Per Eriksson
- grid.5640.70000 0001 2162 9922Department of Biomedical and Clinical Sciences, Division of Inflammation and Infection, Linköping University, Linköping, Sweden
| | - Simon J. Bowman
- grid.412563.70000 0004 0376 6589Rheumatology Department, University Hospital Birmingham NHS Foundation Trust, Birmingham, UK ,grid.6572.60000 0004 1936 7486Rheumatology Research Group, Institute of Inflammation & Ageing, University of Birmingham, Birmingham, UK ,grid.415667.7Rheumatology Department, Milton Keynes University Hospital, Milton Keynes, UK
| | - Roald Omdal
- grid.7914.b0000 0004 1936 7443Department of Clinical Science, University of Bergen, Bergen, Norway ,grid.412835.90000 0004 0627 2891Department of Internal Medicine, Clinical Immunology Unit, Stavanger University Hospital, Stavanger, Norway
| | - Lars Rönnblom
- grid.8993.b0000 0004 1936 9457Department of Medical Sciences, Rheumatology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Blake Warner
- grid.419633.a0000 0001 2205 0568Salivary Disorder Unit, National Institute of Dental and Craniofacial Research, Bethesda, MD USA
| | - Maureen Rischmueller
- grid.278859.90000 0004 0486 659XRheumatology Department, The Queen Elizabeth Hospital, Woodville, South Australia ,grid.1010.00000 0004 1936 7304University of Adelaide, Adelaide, South Australia
| | - Torsten Witte
- grid.10423.340000 0000 9529 9877Department of Rheumatology and Immunology, Hannover Medical School, Hannover, Germany
| | - A. Darise Farris
- grid.274264.10000 0000 8527 6890Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK USA
| | - Xavier Mariette
- grid.413784.d0000 0001 2181 7253Université Paris-Saclay, Assistance Publique–Hôpitaux de Paris (AP-HP), Hôpital Bicêtre, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1184, Le Kremlin Bicêtre, France
| | - Marta E. Alarcon-Riquelme
- grid.4489.10000000121678994Genyo, Center for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, Granada, Spain
| | | | - Caroline H. Shiboski
- grid.266102.10000 0001 2297 6811Department of Orofacial Sciences, University of California San Francisco, San Francisco, CA USA
| | | | - Marie Wahren-Herlenius
- grid.7914.b0000 0004 1936 7443Department of Clinical Science, University of Bergen, Bergen, Norway ,grid.4714.60000 0004 1937 0626Rheumatology Unity, Department of Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Wan-Fai Ng
- grid.1006.70000 0001 0462 7212Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK ,grid.420004.20000 0004 0444 2244NIHR Newcastle Biomedical Centre and NIHR Newcastle Clinical Research Facility, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | | | - Kathy L. Sivils
- grid.274264.10000 0000 8527 6890Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK USA ,grid.505430.7Translational Sciences, The Janssen Pharmaceutical Companies of Johnson & Johnson, Spring House, PA USA
| | - Indra Adrianto
- grid.239864.20000 0000 8523 7701Center for Bioinformatics, Department of Public Health Sciences, Henry Ford Health System, Detroit, MI USA
| | - Gunnel Nordmark
- grid.8993.b0000 0004 1936 9457Department of Medical Sciences, Rheumatology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Christopher J. Lessard
- grid.274264.10000 0000 8527 6890Genes and Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK USA ,grid.266902.90000 0001 2179 3618Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
| |
Collapse
|
19
|
Patel V, Ma S, Yadlapati R. Salivary biomarkers and esophageal disorders. Dis Esophagus 2022; 35:6566015. [PMID: 35397479 DOI: 10.1093/dote/doac018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 03/15/2022] [Indexed: 12/11/2022]
Abstract
Saliva is a complex physiologic fluid that contains an abundance of biological analytes, or biomarkers. Recent research has shown that these biomarkers may be able to convey the physiologic health of a person. Work has been done linking derangements in these salivary biomarkers to a wide variety of pathologic disorders ranging from oncologic diseases to atopic conditions. The specific area of interest for this review paper is esophageal disorders. Particularly because the diagnosis and management of esophageal disorders often includes invasive testing such as esophagogastroduodenoscopy, prolonged pH monitoring, and biopsy. The aim of this review will be to explore salivary biomarkers (pepsin, bile, epidermal growth factor, and micro-RNA) that are being studied as they relate specifically to esophageal disorders. Finally, it will explore the benefits of salivary testing and identify areas of possible future research.
Collapse
Affiliation(s)
- Vandan Patel
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Steven Ma
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Rena Yadlapati
- Division of Gastroenterology, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
20
|
Karimi B, Dehghani Firoozabadi A, Peymani M, Ghaedi K. Circulating long noncoding RNAs as novel bio-tools: Focus on autoimmune diseases. Hum Immunol 2022; 83:618-627. [PMID: 35717260 DOI: 10.1016/j.humimm.2022.06.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 06/04/2022] [Accepted: 06/07/2022] [Indexed: 11/04/2022]
Abstract
Long non-coding RNAs (lncRNAs) are an emerging class of non-coding RNAs that do not encode proteins. These RNAs have various essential regulatory functions. Irregular expression of lncRNAs has been related to the pathological process of varied diseases, and are considered promising diagnostic biomarkers. LncRNAs can release into the circulation and be stable in body fluids as circulating lncRNAs. A subset of circulating lncRNAs that exist in exosomes are referred to as exosomal lncRNA molecules. These lncRNAs are highly stable and resist RNases. Exosomes have captured a great deal of attention due to their involvement in regulating communications between cells. In conditions of autoimmune disease, exosomes play critical roles in the pathological processes. In this context, circulating lncRNAs have been shown to modulate the immune response and indicated as prognosis and diagnostic biomarkers for autoimmune diseases. This review highlights the role of circulating lncRNAs (particularly exosomal) as diagnostic biomarkers for autoimmune diseases such as rheumatoid arthritis, systemic lupus erythematosus, multiple sclerosis, psoriasis, and Sjögren's syndrome.
Collapse
Affiliation(s)
- Bahareh Karimi
- Department of Cellular and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | | | - Maryam Peymani
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Kamran Ghaedi
- Department of Cellular and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran.
| |
Collapse
|
21
|
Pultrone L, Schmid R, Waltimo T, Braissant O, Astasov-Frauenhoffer M. Saliva profiling with differential scanning calorimetry: A feasibility study with ex vivo samples. PLoS One 2022; 17:e0269600. [PMID: 35687571 PMCID: PMC9187081 DOI: 10.1371/journal.pone.0269600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 05/24/2022] [Indexed: 11/18/2022] Open
Abstract
Differential scanning calorimetry (DSC) has been used widely to study various biomarkers from blood, less is known about the protein profiles from saliva. The aim of the study was to investigate the use DSC in order to detect saliva thermal profiles and determine the most appropriate sampling procedure to collect and process saliva. Saliva was collected from 25 healthy young individuals and processed using different protocols based on centrifugation and filtering. The most effective protocol was centrifugation at 5000g for 10 min at 4°C followed by filtration through Millex 0.45 μm filter. Prepared samples were transferred to 3 mL calorimetric ampoules and then loaded into TAM48 calibrated to 30°C until analysis. DSC scans were recorded from 30°C to 90°C at a scan rate of 1°C/h with a pre-conditioning the samples to starting temperature for 1 h. The results show that the peak distribution of protein melting points was clearly bimodal, and the majority of peaks appeared between 40–50°C. Another set of peaks is visible between 65°C– 75°C. Additionally, the peak amplitude and area under the peak are less affected by the concentration of protein in the sample than by the individual differences between people. In conclusion, the study shows that with right preparation of the samples, there is a possibility to have thermograms of salivary proteins that show peaks in similar temperature regions between different healthy volunteers.
Collapse
Affiliation(s)
- Lena Pultrone
- Clinic for Oral Health & Medicine, University Center for Dental Medicine Basel UZB, University of Basel, Basel, Switzerland
| | - Raphael Schmid
- Clinic for Oral Health & Medicine, University Center for Dental Medicine Basel UZB, University of Basel, Basel, Switzerland
| | - Tuomas Waltimo
- Clinic for Oral Health & Medicine, University Center for Dental Medicine Basel UZB, University of Basel, Basel, Switzerland
| | - Olivier Braissant
- Center of Biomechanics and Biocalorimetry, c/o Department of Biomedical Engineering (DBE), University of Basel, Allschwil, Switzerland
| | - Monika Astasov-Frauenhoffer
- Department Research, University Center for Dental Medicine Basel UZB, University of Basel, Basel, Switzerland
- * E-mail:
| |
Collapse
|
22
|
Li Z, Mu Y, Guo C, You X, Liu X, Li Q, Sun W. Analysis of the saliva metabolic signature in patients with primary Sjögren’s syndrome. PLoS One 2022; 17:e0269275. [PMID: 35653354 PMCID: PMC9162338 DOI: 10.1371/journal.pone.0269275] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 05/17/2022] [Indexed: 11/18/2022] Open
Abstract
Background
The saliva metabolome has been applied to explore disease biomarkers. In this study we characterized the metabolic profile of primary Sjögren’s syndrome (pSS) patients and explored metabolomic biomarkers.
Methods
This work presents a liquid chromatography-mass spectrometry-based metabolomic study of the saliva of 32 patients with pSS and 38 age- and sex-matched healthy adults. Potential pSS saliva metabolite biomarkers were explored using test group saliva samples (20 patients with pSS vs. 25 healthy adults) and were then verified by a cross-validation group (12 patients with pSS vs. 13 healthy adults).
Results
Metabolic pathways, including tryptophan metabolism, tyrosine metabolism, carbon fixation, and aspartate and asparagine metabolism, were found to be significantly regulated and related to inflammatory injury, neurological cognitive impairment and the immune response. Phenylalanyl-alanine was discovered to have good predictive ability for pSS, with an area under the curve (AUC) of 0.87 in the testing group (validation group: AUC = 0.75).
Conclusion
Our study shows that salivary metabolomics is a useful strategy for differential analysis and biomarker discovery in pSS.
Collapse
Affiliation(s)
- Zhen Li
- Department of Stomatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Yue Mu
- Department of Stomatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Chunlan Guo
- Department of Stomatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Xin You
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaoyan Liu
- Core Facility of Instrument, Chinese Academy of Medical Sciences, School of Basic Medicine, Institute of Basic Medical Sciences, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Qian Li
- Department of Stomatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
- * E-mail: (QL); (WS)
| | - Wei Sun
- Core Facility of Instrument, Chinese Academy of Medical Sciences, School of Basic Medicine, Institute of Basic Medical Sciences, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
- * E-mail: (QL); (WS)
| |
Collapse
|
23
|
Mo YQ, Nakamura H, Tanaka T, Odani T, Perez P, Ji Y, French BN, Pranzatelli TJ, Michael DG, Yin H, Chow SS, Khalaj M, Afione SA, Zheng C, Oliveira FR, Motta ACF, Ribeiro-Silva A, Rocha EM, Nguyen CQ, Noguchi M, Atsumi T, Warner BM, Chiorini JA. Lysosomal exocytosis of HSP70 stimulates monocytic BMP6 expression in Sjögren's syndrome. J Clin Invest 2022; 132:e152780. [PMID: 35113815 PMCID: PMC8920330 DOI: 10.1172/jci152780] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 01/28/2022] [Indexed: 12/04/2022] Open
Abstract
BMP6 is a central cytokine in the induction of Sjögren's syndrome-associated (SS-associated) secretory hypofunction. However, the upstream initiation leading to the production of this cytokine in SS is unknown. In this study, RNA ISH on salivary gland sections taken from patients with SS indicated monocytic lineage cells as a cellular source of BMP6. RNA-Seq data on human salivary glands suggested that TLR4 signaling was an upstream regulator of BMP6, which was confirmed by in vitro cell assays and single-cell transcriptomics of human PBMCs. Further investigation showed that HSP70 was an endogenous natural TLR4 ligand that stimulated BMP6 expression in SS. Release of HSP70 from epithelial cells could be triggered by overexpression of lysosome-associated membrane protein 3 (LAMP3), a protein also associated with SS in several transcriptome studies. In vitro studies supported the idea that HSP70 was released as a result of lysosomal exocytosis initiated by LAMP3 expression, and reverse transcription PCR on RNA from minor salivary glands of patients with SS confirmed a positive correlation between BMP6 and LAMP3 expression. BMP6 expression could be experimentally induced in mice by overexpression of LAMP3, which developed an SS-like phenotype. The newly identified LAMP3/HSP70/BMP6 axis provided an etiological model for SS gland dysfunction and autoimmunity.
Collapse
Affiliation(s)
| | | | | | | | - Paola Perez
- AAV Biology Section and
- Salivary Disorder Unit, National Institute of Dental and Craniofacial Research, NIH, Bethesda, Maryland, USA
| | | | | | | | | | | | | | | | | | | | | | - Ana Carolina F. Motta
- Department of Stomatology, Public Health and Forensic Dentistry, School of Dentistry of Ribeirão Preto
| | | | - Eduardo M. Rocha
- Department of Ophthalmology, Otorhinolaryngology, Head and Neck Surgery, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Cuong Q. Nguyen
- Department of Pathology and Infectious Diseases, University of Florida, Gainesville, Florida, USA
| | | | - Tatsuya Atsumi
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Blake M. Warner
- AAV Biology Section and
- Salivary Disorder Unit, National Institute of Dental and Craniofacial Research, NIH, Bethesda, Maryland, USA
| | | |
Collapse
|
24
|
Costa-da-Silva AC, Aure MH, Dodge J, Martin D, Dhamala S, Cho M, Rose JJ, Bassim CW, Ambatipudi K, Hakim FT, Pavletic SZ, Mays JW. Salivary ZG16B expression loss follows exocrine gland dysfunction related to oral chronic graft-versus-host disease. iScience 2022; 25:103592. [PMID: 35005541 PMCID: PMC8718990 DOI: 10.1016/j.isci.2021.103592] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 11/01/2021] [Accepted: 12/06/2021] [Indexed: 11/15/2022] Open
Abstract
Chronic graft-versus-host disease (cGVHD) targets include the oral mucosa and salivary glands after allogeneic hematopoietic stem cell transplant (HSCT). Without incisional biopsy, no diagnostic test exists to confirm oral cGVHD. Consequently, therapy is often withheld until severe manifestations develop. This proteomic study examined saliva and human salivary gland for a biomarker profile at first onset of oral cGVHD prior to initiation of topical steroid therapy. Whole saliva collected at onset of biopsy-proven oral GVHD was assessed using liquid chromatography-coupled tandem mass spectrometry with identification of 569 proteins, of which 77 significantly changed in abundance. ZG16B, a secretory lectin protein, was reduced 2-fold in oral cGVHD saliva (p <0.05), and significantly decreased in salivary gland secretory cells affected by cGVHD. Single-cell RNA-seq analysis of healthy MSG localized ZG16B expression to two discrete acinar cell populations. Reduced ZG16B expression may indicate specific cGVHD activity and possibly general salivary gland dysfunction.
Collapse
Affiliation(s)
- Ana Caroline Costa-da-Silva
- National Institute of Dental and Craniofacial Research, NIH, Building 30, MSC 4340, 30 Convent Drive, Bethesda, MD 20892, USA
| | - Marit H. Aure
- National Institute of Dental and Craniofacial Research, NIH, Building 30, MSC 4340, 30 Convent Drive, Bethesda, MD 20892, USA
| | - Joshua Dodge
- National Institute of Dental and Craniofacial Research, NIH, Building 30, MSC 4340, 30 Convent Drive, Bethesda, MD 20892, USA
| | - Daniel Martin
- National Institute of Dental and Craniofacial Research, NIH, Building 30, MSC 4340, 30 Convent Drive, Bethesda, MD 20892, USA
| | - Susan Dhamala
- National Institute of Dental and Craniofacial Research, NIH, Building 30, MSC 4340, 30 Convent Drive, Bethesda, MD 20892, USA
- National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Monica Cho
- National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | | | - Carol W. Bassim
- National Institute of Dental and Craniofacial Research, NIH, Building 30, MSC 4340, 30 Convent Drive, Bethesda, MD 20892, USA
| | - Kiran Ambatipudi
- National Institute of Dental and Craniofacial Research, NIH, Building 30, MSC 4340, 30 Convent Drive, Bethesda, MD 20892, USA
| | | | | | - Jacqueline W. Mays
- National Institute of Dental and Craniofacial Research, NIH, Building 30, MSC 4340, 30 Convent Drive, Bethesda, MD 20892, USA
| |
Collapse
|
25
|
AlJasser R, AlAqeely R, AlKenani M, AlQahtani S, AlZahrani A, Lambarte R. The effect of systemic Isotretinoin on salivary tissue inhibitors of metalloproteinases 1 and 2 and salivary flow rate in periodontal disease. Saudi J Biol Sci 2022; 29:148-153. [PMID: 35002402 PMCID: PMC8716862 DOI: 10.1016/j.sjbs.2021.08.079] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 08/11/2021] [Accepted: 08/22/2021] [Indexed: 10/31/2022] Open
Abstract
AIMS To evaluate and compare changes in salivary flow rate and salivary levels of TIMP-1 and TIMP-2 in individuals taking oral Isotretinoin (INN) with those who do not take INN. To assess the variation in TIMP-1 and TIMP-2 as well as salivary flow rate observed at different stages of periodontal disease in comparison to those observed in the case of healthy periodontium. MATERIALS AND METHODS An examiner-blind case-control study involving 180 human adults divided into six groups based on their periodontal status. Clinical parameters, including pocket depth, clinical attachment level, and bleeding on probing were measured at six sites per tooth. Whole unstimulated saliva samples were collected from all subjects to evaluate salivary flow rate (SFR). Salivary TIMP-1 and TIMP-2 levels were detected using enzyme-linked immunosorbent assay (ELISA). Data were analyzed using IBM SPSS Software. The Kruskal Wallis test and Mann-Whitney U-tests were employed to verify any significant differences between the groups for all parameters. Multi-regression analysis was performed for each parameter tested in each group. All tests were compared at a significance level of 0.05. RESULTS SFR was statistically significantly lower among all INN groups in comparison to the control groups (P < 0.001). TIMP-1 and TIMP-2 were significantly higher in all INN groups in comparison to the control groups, in both gingivitis cases (P = 0.004, P < 0.0001 respectively) and periodontitis cases (P < 0.0001). CONCLUSION Although INN reduces salivary flow rate, the findings of the present study revealed that it had an anti-inflammatory effect in periodontal biomarkers. Specifically, it was positively correlated with an elevation of salivary TIMP-1 and TIMP-2. Hence, INN might be a future additive medication to be further evaluated for the treatment of periodontal diseases.
Collapse
Affiliation(s)
- Reham AlJasser
- Department of Periodontics and Community Dentistry, Dental College, King Saud University, 11545 Riyadh, Saudi Arabia, Arabia
| | - Razan AlAqeely
- Department of Periodontics and Community Dentistry, Dental College, King Saud University, 11545 Riyadh, Saudi Arabia, Arabia
| | - Manal AlKenani
- Saudi Board of Periodontics Program, 12211 Riyadh, Saudi Arabia
| | | | | | - Rhodanne Lambarte
- Mollecular and Cell Biology Laboratory Prince Naif bin Abdul Aziz Health Research Center, College of Dentistry, Dental College, King Saud University, 11545 Riyadh, Saudi Arabia, Arabia
| |
Collapse
|
26
|
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
|
27
|
Pontarini E, Coleby R, Bombardieri M. Cellular and molecular diversity in Sjogren's syndrome salivary glands: Towards a better definition of disease subsets. Semin Immunol 2021; 58:101547. [PMID: 34876330 DOI: 10.1016/j.smim.2021.101547] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Primary Sjögren's syndrome (pSS) is a highly heterogeneous disease in terms of clinical presentation ranging from a mild disease localised to the salivary and lacrimal glands, to multiorgan complications of various degrees of severity, finishing with the evolution, in around 5% of pSS patients, to B cell lymphomas most commonly arising in the inflamed salivary glands. Currently, there are poor positive or negative predictors of disease evolution able to guide patient management and treatment at early stages of the diseases. Recent understanding of the pathogenic mechanisms driving immunopathology in pSS, particularly through histological and transcriptomic analysis of minor and parotid salivary gland (SG) biopsies, has highlighted a high degree of cellular and molecular heterogeneity of the inflammatory lesions but also allowed the identification of clusters of patients with similar underlying SG immunopathology. In particular, patients presenting with high degrees of B/T cell infiltration and the formation of ectopic lymphoid structures (ELS) in the SG have been associated, albeit with conflicting results, with higher degree of disease severity and enhanced risk of lymphoma evolution, suggesting that a dysregulated adaptive immune response plays a key role in driving disease manifestations in pSS. Recent data from randomised clinical trials with novel biological therapies in pSS have also highlighted the potential role of SG immunopathology and molecular pathology in stratifying patients for trial inclusion as well as assessing proof of mechanisms in longitudinal SG biopsies before and after treatment. Although significant progress has been made in the understanding of disease pathogenesis and heterogeneity through cellular and molecular SG pathology, further work is needed to validate their clinical utility in routine clinical settings and in randomised clinical trials.
Collapse
Affiliation(s)
- Elena Pontarini
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Rachel Coleby
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Michele Bombardieri
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom.
| |
Collapse
|
28
|
Jung JY, Kim JW, Kim HA, Suh CH. Salivary Biomarkers in Patients with Sjögren's Syndrome-A Systematic Review. Int J Mol Sci 2021; 22:12903. [PMID: 34884709 PMCID: PMC8657642 DOI: 10.3390/ijms222312903] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 11/24/2021] [Accepted: 11/26/2021] [Indexed: 12/18/2022] Open
Abstract
Sjögren's syndrome (SS) is a chronic autoimmune disease characterized by dry mouth and dry eyes, with lymphocytic infiltration of the exocrine glands. Saliva is becoming a useful tool to determine the clinical and pathological characteristics of SS because the collection method is easy and non-invasive. Since 1900, salivary proteomic analysis has been performed continuously using a variety of optimized analytical methods. Many studies have identified distinct characteristics of salivary proteins in patients with primary SS, and the changes were related to chronic inflammation and overproduction of immunoglobulins or downregulated secretory function. Several proteomic studies using whole or parotid saliva have evaluated whether several salivary proteins can be used to discriminate SS, including salivary β2-microglobulin, calprotectin, carbonic anhydrase VI, neutrophil gelatinase-associated lipocalin, sialic acid-binding immunoglobulin-like lectin-5, and tripartite motif-containing protein 29. In addition, salivary proinflammatory cytokine levels have been reported to be increased in patients with SS. Although these candidate salivary proteins have exhibited considerable differences in patients with SS, more data are needed to confirm their role as biomarkers. Moreover, the identification of salivary characteristics that can accurately reflect disease activity, predict treatment response and prognosis, and diagnose SS is anticipated.
Collapse
Affiliation(s)
| | | | | | - Chang-Hee Suh
- Department of Rheumatology, Ajou University School of Medicine, Suwon 16499, Korea; (J.-Y.J.); (J.-W.K.); (H.-A.K.)
| |
Collapse
|
29
|
Qi X, Wang XQ, Jin L, Gao LX, Guo HF. Uncovering potential single nucleotide polymorphisms, copy number variations and related signaling pathways in primary Sjogren's syndrome. Bioengineered 2021; 12:9313-9331. [PMID: 34723755 PMCID: PMC8809958 DOI: 10.1080/21655979.2021.2000245] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Primary Sjogren’s syndrome (pSS) is a complex systemic autoimmune disease, which is difficult to accurately diagnose due to symptom diversity in patients, especially at earlier stages. We tried to find potential single nucleotide polymorphisms (SNPs), copy number variations (CNVs) and related signaling pathways. Genomic DNA was extracted from peripheral blood of 12 individuals (7 individuals from 3 pSS pedigrees and 5 sporadic cases) for whole-exome sequencing (WES) analysis. SNPs and CNVs were identified, followed by functional annotation of genes with SNPs and CNVs. Gene expression profile (involving 64 normal controls and 166 cases) was downloaded from the Gene Expression Omnibus database (GEO) dataset for differentially expression analysis. Sanger sequencing and in vitro validation was used to validate the identified SNPs and differentially expressed genes, respectively. A total of 5 SNPs were identified in both pedigrees and sporadic cases, such as FES, PPM1J, and TRAPPC9. A total of 3402 and 19 CNVs were identified in pedigrees and sporadic cases, respectively. Fifty-one differentially expressed genes were associated with immunity, such as BATF3, LAP3, BATF2, PARP9, and IL15RA. AMPK signaling pathway and cell adhesion molecules (CAMs) were the most significantly enriched signaling pathways of identified SNPs. Identified CNVs were associated with systemic lupus erythematosus, mineral absorption, and HTLV-I infection. IL2-STAT5 signaling, interferon-gamma response, and interferon-alpha response were significantly enriched immune related signaling pathways of identified differentially expressed genes. In conclusion, our study found some potential SNPs, CNVs, and related signaling pathways, which could be useful in understanding the pathological mechanism of pSS.
Collapse
Affiliation(s)
- Xuan Qi
- Department of Rheumatism and Immunology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xi-Qin Wang
- Internal Medicine, Yuhua Yunfang Integrated Traditional Chinese and Western Medicine Clinic, Shijiazhuang, Hebei, China
| | - Lu Jin
- Department of Rheumatism and Immunology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Li-Xia Gao
- Department of Rheumatism and Immunology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Hui-Fang Guo
- Department of Rheumatism and Immunology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
30
|
AlJasser R, Zahid M, AlSarhan M, AlOtaibi D, AlOraini S. The effect of conventional versus electronic cigarette use on treatment outcomes of peri-implant disease. BMC Oral Health 2021; 21:480. [PMID: 34579704 PMCID: PMC8477469 DOI: 10.1186/s12903-021-01784-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 08/25/2021] [Indexed: 11/10/2022] Open
Abstract
Aim To compare changes in clinical periodontal parameters (gingival consistency, colour, BOP, PI, PD) and changes of salivary inflammatory biomarkers (IL-1 β, IL-6, MMP-8, TNF- α and TIMP-1 between conventional, electronic cigarette smokers and non-smokers after peri-implant treatment. Methods Study participants were grouped into three groups (i) Conventional cigarette smokers (ii) Electronic cigarette smokers and (iii) non-smokers respectively. A total of 60 adult patients aged (40–56 years) with 60 implants with active per-impantitis was included.Clinical and Biological parameters were evaluated before surgical treatment at baseline, one, six and twelve month post treatment. Pearson’s chi-square test was used to compare the distribution of the categorical while Two-way repeated analysis of variance was used to compare the mean values of quantitative outcome variables among all study groups across the 4 time points. Results A total of 60 subjects (60 implants) were selected and classified into three groups as per their smoking method 20 participants in each group with one single targeted implant diagnosis with active peri-implantitis. The gingival colour, the change was statistically significant at one year of post treatment.The gingival consistency distribution across the three groups is not statistically significant at baseline, but it is statistically significant at one-month (p = 0.001), six months (p = 0.029) and at the completion of one-year (p = 0.018) post treatment. The plaque index of 100% of non-smokers had changed to ‘0’ and 35% change in cigarettes and 30% change in electronic smokers which is statistically significant (p = 0.016).The prevalence of BOP was observed in the three groups as 72%, 76.5% and 88.9% at baseline. The mean values of PD have shown statistically significant change across the three groups over the four-time intervals of observation (p = 0.024). The comparison of mean values of IL-1 β, IL-6 and TIMP-1 has shown statistically significant change across the three groups over the four intervals of observation (p < 0.0001). Conclusions Electronic cigarette smoking was found to be most prevalent risk indicator for peri-implantitis. Compromised response of peri-implantitis treatment both clinically and biologically was found more among electronic cigarette smokers when compared to conventional cigarette smokers and non-smokers. Trial registration: This case-control study was conducted at King Saud University’s Dental College, Riyadh, Saudi Arabia, in accordance with “Helsinki Declaration of Human Studies” and approved by the Institutional Review Board (Reference no: 87563).
Collapse
Affiliation(s)
- Reham AlJasser
- Department of Periodontics and Community Dentistry, Dental College, King Saud University, PO Box 60169, Riyadh, 11545, Saudi Arabia.
| | - Mohammed Zahid
- Saudi Board of Periodontics Resident, Ministry of Health, Riyadh, Saudi Arabia
| | - Mohammed AlSarhan
- Department of Periodontics and Community Dentistry, Dental College, King Saud University, PO Box 60169, Riyadh, 11545, Saudi Arabia
| | - Dalal AlOtaibi
- Department of Periodontics and Community Dentistry, Dental College, King Saud University, PO Box 60169, Riyadh, 11545, Saudi Arabia
| | - Saleh AlOraini
- Department of Periodontics and Community Dentistry, Dental College, King Saud University, PO Box 60169, Riyadh, 11545, Saudi Arabia
| |
Collapse
|
31
|
de Paiva CS, Trujillo-Vargas CM, Schaefer L, Yu Z, Britton RA, Pflugfelder SC. Differentially Expressed Gene Pathways in the Conjunctiva of Sjögren Syndrome Keratoconjunctivitis Sicca. Front Immunol 2021; 12:702755. [PMID: 34349764 PMCID: PMC8326832 DOI: 10.3389/fimmu.2021.702755] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 07/01/2021] [Indexed: 12/24/2022] Open
Abstract
Sjögren syndrome (SS) is an autoimmune condition that targets the salivary and lacrimal glands, with cardinal clinical signs of dry eye (keratoconjunctivitis sicca, KCS) and dry mouth. The conjunctiva of SS patients is often infiltrated by immune cells that participate in the induction and maintenance of local inflammation. The purpose of this study was to investigate immune-related molecular pathways activated in the conjunctiva of SS patients. Female SS patients (n=7) and controls (n=19) completed a series of oral, ocular surface exams. Symptom severity scores were evaluated using validated questionnaires (OSDI and SANDE). All patients fulfilled the ACR/EULAR criteria for SS and the criteria for KCS. Fluorescein and lissamine green dye staining evaluated tear-break-up time (TBUT), corneal and conjunctival disease, respectively. Impression cytology of the temporal bulbar conjunctiva was performed to collect cells lysed and subjected to gene expression analysis using the NanoString Immunology Panel. 53/594 differentially expressed genes (DEGs) were observed between SS and healthy controls; 49 DEGs were upregulated, and 4 were downregulated (TRAF5, TGFBI, KLRAP1, and CMKLRI). The top 10 DEGs in descending order were BST2, IFITM1, LAMP3, CXCL1, IL19, CFB, LY96, MX1, IL4R, CDKN1A. Twenty pathways had a global significance score greater or equal to 2. Spearman correlations showed that 29/49 upregulated DEGs correlated with either TBUT (inverse) or OSDI or conjunctival staining score (positive correlations). Venn diagrams identified that 26/29 DEGs correlated with TBUT, 5/26 DEGs correlated with OSDI, and 16/26 correlated with conjunctival staining scores. Five upregulated DEGs (CFB, CFI, IL1R1, IL2RG, IL4R) were uniquely negatively correlated with TBUT. These data indicate that the conjunctiva of SS patients exhibits a phenotype of immune activation, although some genes could be inhibitory. Some of the DEGs and pathways overlap with previous DEGs in salivary gland biopsies, but new DEGs were identified, and some of these correlated with symptoms and signs of dry eye. Our results indicate that gene analysis of conjunctiva imprints is a powerful tool to understand the pathogenesis of SS and develop new therapeutic targets.
Collapse
Affiliation(s)
- Cintia S. de Paiva
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX, United States
| | - Claudia M. Trujillo-Vargas
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX, United States
- Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
- Grupo de Inmunodeficiencias Primarias, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín, Colombia
| | - Laura Schaefer
- Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - Zhiyuan Yu
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX, United States
| | - Robert A. Britton
- Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | | |
Collapse
|
32
|
The Potential Impact of Salivary IL-1 on the Diagnosis of Periodontal Disease: A Pilot Study. Healthcare (Basel) 2021; 9:healthcare9060729. [PMID: 34199256 PMCID: PMC8231867 DOI: 10.3390/healthcare9060729] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/11/2021] [Accepted: 06/11/2021] [Indexed: 12/29/2022] Open
Abstract
The aim of this study was to identify inflammatory cytokines as salivary biomarkers for periodontal disease. The subjects were 33 Korean adults aged 23 to 71 years. Using a multiplexed bead immunoassay called Luminex, the levels of inflammatory cytokines related to periodontal disease were evaluated. Oral examination for periodontal disease and gingival bleeding was conducted. With these two independent variables, differences in inflammatory cytokines were analyzed by an independent t-test and age-adjusted ANCOVA. Among the subjects, 21 had periodontal disease and 12 were healthy subjects. The gingival bleeding status was classified into low and high levels. Among 13 inflammatory cytokines in saliva, IL-1α, IL-1β, IL-4, IL-8, CCL2/MCP-1, CCL3/MIP-1α, and TNF-α were found to be significant biomarkers within the standard curve. The quantity of IL-1β was increased in subjects with high levels of gingival bleeding. IL-1α levels were increased in subjects with periodontal disease. After adjusting for age, the significant biomarkers for gingival bleeding and periodontal disease were IL-1β and IL-1α, respectively. Using the receiver operating characteristic (ROC) curve, IL-1β was confirmed as a significant biomarker. The sensitivity and specificity of IL-1β for predicting periodontitis were 88.24% and 62.5%, respectively. Therefore, IL-1 was found to be a significant biomarker for periodontal disease, and it could be used in the diagnosis of periodontal disease using saliva.
Collapse
|
33
|
Pin E, Petricoin EF, Cortes N, Bowman TG, Andersson E, Uhlén M, Nilsson P, Caswell SV. Immunoglobulin A Autoreactivity toward Brain Enriched and Apoptosis-Regulating Proteins in Saliva of Athletes after Acute Concussion and Subconcussive Impacts. J Neurotrauma 2021; 38:2373-2383. [PMID: 33858214 DOI: 10.1089/neu.2020.7375] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The diagnosis and management of concussion is hindered by its diverse clinical presentation and assessment tools reliant on subjectively experienced symptoms. The biomechanical threshold of concussion is also not well understood, and asymptomatic concussion or "subconcussive impacts" of variable magnitudes are common in contact sports. Concerns have risen because athletes returning to activity too soon have an increased risk of prolonged recovery or long-term adverse health consequences. To date, little is understood on a molecular level regarding concussion and subconcussive impacts. Recent research suggests that neuroinflammatory mechanisms may serve an important role subsequent to concussion and possibly to subconcussive impacts. These studies suggest that autoantibodies may be a valuable tool for detection of acute concussion and monitoring for changes caused by cumulative exposure to subconcussive impacts. Hence, we aimed to profile the immunoglobulin (Ig)A autoantibody repertoire in saliva by screening a unique sport-related head trauma biobank. Saliva samples (n = 167) were donated by male and female participants enrolled in either the concussion (24-48 h post-injury) or subconcussion (non-concussed participants having moderate or high cumulative subconcussive impact exposure) cohorts. Study design included discovery and verification phases. Discovery aimed to identify new candidate autoimmune targets of IgA. Verification tested whether concussion and subconcussion cohorts increased IgA reactivity and whether cohorts showed similarities. The results show a significant increase in the prevalence of IgA toward protein fragments representing 5-hydroxytryptamine receptor 1A (HTR1A), serine/arginine repetitive matrix 4 (SRRM4) and FAS (tumor necrosis factor receptor superfamily member 6) after concussion and subconcussion. These results may suggest that concussion and subconcussion induce similar physiological effects, especially in terms of immune response. Our study demonstrates that saliva is a potential biofluid for autoantibody detection in concussion and subconcussion. After rigorous confirmation in much larger independent study sets, a validated salivary autoantibody assay could provide a non-subjective quantitative means of assessing concussive and subconcussive events.
Collapse
Affiliation(s)
- Elisa Pin
- Division of Affinity Proteomics, Department of Protein Science, Department of Protein Science, KTH Royal Institute of Technology, SciLifeLab, Stockholm, Sweden
| | - Emanuel F Petricoin
- Center for Applied Proteomics and Molecular Medicine, School of Kinesiology, George Mason University, Manassas, Virginia, USA.,Institute for BioHealth Innovation, and School of Kinesiology, George Mason University, Manassas, Virginia, USA
| | - Nelson Cortes
- Institute for BioHealth Innovation, and School of Kinesiology, George Mason University, Manassas, Virginia, USA.,Sports Medicine Assessment Research and Testing Laboratory, School of Kinesiology, George Mason University, Manassas, Virginia, USA
| | - Thomas G Bowman
- Department of Athletic Training, University of Lynchburg, Lynchburg, Virginia, USA
| | - Eni Andersson
- Division of Affinity Proteomics, Department of Protein Science, Department of Protein Science, KTH Royal Institute of Technology, SciLifeLab, Stockholm, Sweden
| | - Mathias Uhlén
- Division of Systems Biology, Department of Protein Science, KTH Royal Institute of Technology, SciLifeLab, Stockholm, Sweden
| | - Peter Nilsson
- Division of Affinity Proteomics, Department of Protein Science, Department of Protein Science, KTH Royal Institute of Technology, SciLifeLab, Stockholm, Sweden
| | - Shane V Caswell
- Institute for BioHealth Innovation, and School of Kinesiology, George Mason University, Manassas, Virginia, USA.,Sports Medicine Assessment Research and Testing Laboratory, School of Kinesiology, George Mason University, Manassas, Virginia, USA
| |
Collapse
|
34
|
Xu H, Chen J, Wang Y, Wu Y, Liang Y. SELL and IFI44 as potential biomarkers of Sjögren's syndrome and their correlation with immune cell infiltration. Genes Genet Syst 2021; 96:71-80. [PMID: 33883324 DOI: 10.1266/ggs.20-00053] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The onset of Sjögren's syndrome (SS) is hidden, early diagnosis is difficult, and the disorder seriously endangers the physical and mental health of affected people. This study aims to identify potential biomarkers of SS and to investigate the characteristics of immune cell infiltration. We used four SS gene expression profile data series from the Gene Expression Omnibus database, and applied bioinformatics analysis and machine learning algorithms to screen two biomarkers, SELL (L-selectin) and IFI44 (interferon-induced protein 44), from 101 differentially expressed genes. The two-gene model comprising SELL and IFI44 showed good diagnostic ability for SS in the training set (AUC = 0.992) and verification set (AUC = 0.917). Analysis of infiltrating immune cells in SS identified naive B cells, resting CD4 memory T cells, activated CD4 memory T cells, gamma delta T cells, M0 macrophages, M1 macrophages, plasma cells, CD8 T cells, activated NK cells and monocytes as candidate participants in the SS process. Furthermore, SELL was associated with M2 macrophages, activated CD4 memory T cells, gamma delta T cells, resting NK cells and plasma cells, while IFI44 was associated with activated mast cells, resting NK cells, resting mast cells and CD8 T cells. This study demonstrates that SELL and IFI44 can serve as good diagnostic markers for SS and may also be new diagnostic and therapeutic targets for SS.
Collapse
Affiliation(s)
- Hua Xu
- Department of Laboratory Medicine, Panjin Liaoyou Gem Flower Hospital
| | - Jia Chen
- Department of General Practice, Panjin Liaoyou Gem Flower Hospital
| | - Yang Wang
- Department of Oncology, Panjin Liaoyou Gem Flower Hospital
| | - Yanmei Wu
- Department of Rheumatology and Immunology, Panjin Liaoyou Gem Flower Hospital
| | - Yingjie Liang
- Department of Laboratory Medicine, Panjin Liaoyou Gem Flower Hospital
| |
Collapse
|
35
|
Proctor GB, Shaalan AM. Disease-Induced Changes in Salivary Gland Function and the Composition of Saliva. J Dent Res 2021; 100:1201-1209. [PMID: 33870742 PMCID: PMC8461045 DOI: 10.1177/00220345211004842] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Although the physiological control of salivary secretion has been well studied, the impact of disease on salivary gland function and how this changes the composition and function of saliva is less well understood and is considered in this review. Secretion of saliva is dependent upon nerve-mediated stimuli, which activate glandular fluid and protein secretory mechanisms. The volume of saliva secreted by salivary glands depends upon the frequency and intensity of nerve-mediated stimuli, which increase dramatically with food intake and are subject to facilitatory or inhibitory influences within the central nervous system. Longer-term changes in saliva secretion have been found to occur in response to dietary change and aging, and these physiological influences can alter the composition and function of saliva in the mouth. Salivary gland dysfunction is associated with different diseases, including Sjögren syndrome, sialadenitis, and iatrogenic disease, due to radiotherapy and medications and is usually reported as a loss of secretory volume, which can range in severity. Defining salivary gland dysfunction by measuring salivary flow rates can be difficult since these vary widely in the healthy population. However, saliva can be sampled noninvasively and repeatedly, which facilitates longitudinal studies of subjects, providing a clearer picture of altered function. The application of omics technologies has revealed changes in saliva composition in many systemic diseases, offering disease biomarkers, but these compositional changes may not be related to salivary gland dysfunction. In Sjögren syndrome, there appears to be a change in the rheology of saliva due to altered mucin glycosylation. Analysis of glandular saliva in diseases or therapeutic interventions causing salivary gland inflammation frequently shows increased electrolyte concentrations and increased presence of innate immune proteins, most notably lactoferrin. Altering nerve-mediated signaling of salivary gland secretion contributes to medication-induced dysfunction and may also contribute to altered saliva composition in neurodegenerative disease.
Collapse
Affiliation(s)
- G B Proctor
- Centre for Host Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, UK
| | - A M Shaalan
- Centre for Host Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, UK
| |
Collapse
|
36
|
Yin G, Huang J, Guo W, Huang Z. Metabolomics of Oral/Head and Neck Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1280:277-290. [PMID: 33791989 DOI: 10.1007/978-3-030-51652-9_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/21/2023]
Abstract
Oral/head and neck cancer is the sixth most common human malignancies in the world. Despite the treatment advances in surgery, chemotherapy, and radiotherapy, the patient survival has not been significantly improved in the past several decades. As a new methodological approach, metabolomics may help reveal the metabolic reprogramming mechanisms underlying head and neck cancer cell proliferation, invasion, and metastasis and may be used to identify metabolite biomarkers for clinical applications of the disease. In this chapter, we briefly review recent metabolomic applications in head and neck cancer.
Collapse
Affiliation(s)
- Gaofei Yin
- Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Key Laboratory of Otolaryngology Head and Neck Surgery, Ministry of Education, Capital Medical University, Beijing, China
| | - Junwei Huang
- Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Key Laboratory of Otolaryngology Head and Neck Surgery, Ministry of Education, Capital Medical University, Beijing, China
| | - Wei Guo
- Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Key Laboratory of Otolaryngology Head and Neck Surgery, Ministry of Education, Capital Medical University, Beijing, China
| | - Zhigang Huang
- Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Key Laboratory of Otolaryngology Head and Neck Surgery, Ministry of Education, Capital Medical University, Beijing, China.
| |
Collapse
|
37
|
Belotti Y, Lim CT. Microfluidics for Liquid Biopsies: Recent Advances, Current Challenges, and Future Directions. Anal Chem 2021; 93:4727-4738. [DOI: 10.1021/acs.analchem.1c00410] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Yuri Belotti
- Institute for Health Innovation and Technology, National University of Singapore, 117599 Singapore
| | - Chwee Teck Lim
- Institute for Health Innovation and Technology, National University of Singapore, 117599 Singapore
- Department of Biomedical Engineering, National University of Singapore, 117583 Singapore
- Mechanobiology Institute, National University of Singapore, 117411 Singapore
| |
Collapse
|
38
|
Chatzis L, Vlachoyiannopoulos PG, Tzioufas AG, Goules AV. New frontiers in precision medicine for Sjogren's syndrome. Expert Rev Clin Immunol 2021; 17:127-141. [PMID: 33478279 DOI: 10.1080/1744666x.2021.1879641] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Introduction: Sjögren's syndrome is a unique systemic autoimmune disease, placed in the center of systemic autoimmunity and at the crossroads of autoimmunity and lymphoproliferation. The diverse clinical picture of the disease, the inefficacy of current biologic treatments, and the co-existence with lymphoma conferring to the patients' morbidity and mortality force the scientific community to review disease pathogenesis and reveal the major implicated cellular and molecular elements.Areas covered: Biomarkers for early diagnosis, prediction, stratification, monitoring, and targeted treatments can serve as a tool to interlink and switch from the clinical phenotyping of the disease into a more sophisticated classification based on the underlying critical molecular pathways and endotypes. Such a transition may define the establishment of the so-called precision medicine era in which patients' management will be based on grouping according to pathogenetically related biomarkers. In the current work, literature on Sjogren's syndrome covering several research fields including clinical, translational, and basic research has been reviewed.Expert opinion: The perspectives of clinical and translational research are anticipated to define phenotypic clustering of high-risk pSS patients and link the clinical picture of the disease with fundamental molecular mechanisms and molecules implicated in pathogenesis.
Collapse
Affiliation(s)
- Loukas Chatzis
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Athanasios G Tzioufas
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Andreas V Goules
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
39
|
Huang L, Shao D, Wang Y, Cui X, Li Y, Chen Q, Cui J. Human body-fluid proteome: quantitative profiling and computational prediction. Brief Bioinform 2021; 22:315-333. [PMID: 32020158 PMCID: PMC7820883 DOI: 10.1093/bib/bbz160] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 08/22/2019] [Accepted: 10/18/2019] [Indexed: 12/15/2022] Open
Abstract
Empowered by the advancement of high-throughput bio technologies, recent research on body-fluid proteomes has led to the discoveries of numerous novel disease biomarkers and therapeutic drugs. In the meantime, a tremendous progress in disclosing the body-fluid proteomes was made, resulting in a collection of over 15 000 different proteins detected in major human body fluids. However, common challenges remain with current proteomics technologies about how to effectively handle the large variety of protein modifications in those fluids. To this end, computational effort utilizing statistical and machine-learning approaches has shown early successes in identifying biomarker proteins in specific human diseases. In this article, we first summarized the experimental progresses using a combination of conventional and high-throughput technologies, along with the major discoveries, and focused on current research status of 16 types of body-fluid proteins. Next, the emerging computational work on protein prediction based on support vector machine, ranking algorithm, and protein-protein interaction network were also surveyed, followed by algorithm and application discussion. At last, we discuss additional critical concerns about these topics and close the review by providing future perspectives especially toward the realization of clinical disease biomarker discovery.
Collapse
Affiliation(s)
- Lan Huang
- College of Computer Science and Technology in the Jilin University
| | - Dan Shao
- College of Computer Science and Technology in the Jilin University
- College of Computer Science and Technology in Changchun University
| | - Yan Wang
- College of Computer Science and Technology in the Jilin University
| | - Xueteng Cui
- College of Computer Science and Technology in the Changchun University
| | - Yufei Li
- College of Computer Science and Technology in the Changchun University
| | - Qian Chen
- College of Computer Science and Technology in the Jilin University
| | - Juan Cui
- Department of Computer Science and Engineering in the University of Nebraska-Lincoln
| |
Collapse
|
40
|
Wei P, Xing Y, Li B, Chen F, Hua H. Proteomics-based analysis indicating α-enolase as a potential biomarker in primary Sjögren's syndrome. Gland Surg 2021; 9:2054-2063. [PMID: 33447556 DOI: 10.21037/gs-20-814] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Background Primary Sjögren's syndrome (pSS) is a chronic autoimmune disease. Its etiology is not well understood. Salivary glands are the main target organ in pSS, investigating the changes of salivary protein in pSS patients may not only be a valuable way of identifying new biomarkers/antigens for pSS, but also of revealing the pathogenesis underlying this autoimmune disease. In the present study, we aimed to investigate new biomarkers and explore their potential role in pSS. Methods In this study, α-enolase (ENO1) was found to be overexpressed in pSS by 1D gel electrophoresis/mass spectrometry. The finding was verified by Western blots, immunohistochemistry (IHC), and polymerase chain reaction (PCR) results in both saliva and labial salivary glands. The expression level of immunoglobulin G (IgG) antibody to ENO1 was then tested by enzyme-linked immunosorbent assay (ELISA). Results ENO1 autoantibody was found to be overexpressed in pSS compared with healthy controls. The effects of ENO1 overexpression on rat submandibular gland cell line SMG-C6 was investigated in vitro. The expressions of proteins related to saliva secretion and immunomodulatory were upregulated in ENO1 overexpressed SMG-C6 cells. Conclusions Both ENO1 and anti-ENO1 autoantibody are overexpressed in pSS patients. Nevertheless, their potential role in the pathogenesis of pSS warrants further study.
Collapse
Affiliation(s)
- Pan Wei
- Department of Oral Medicine, Peking University School and Hospital of Stomatology, Beijing, China
| | - Yixiao Xing
- Department of Oral Medicine, Peking University School and Hospital of Stomatology, Beijing, China
| | - Boya Li
- Department of Oral Medicine, Peking University School and Hospital of Stomatology, Beijing, China
| | - Feng Chen
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, China
| | - Hong Hua
- Department of Oral Medicine, Peking University School and Hospital of Stomatology, Beijing, China
| |
Collapse
|
41
|
Huang S, Zheng F, Liu L, Meng S, Cai W, Zhang C, Dai W, Liu D, Hong X, Tang D, Dai Y. Integrated proteome and phosphoproteome analyses of peripheral blood mononuclear cells in primary Sjögren syndrome patients. Aging (Albany NY) 2020; 13:1071-1095. [PMID: 33290261 PMCID: PMC7835054 DOI: 10.18632/aging.202233] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 10/27/2020] [Indexed: 12/18/2022]
Abstract
Primary Sjögren syndrome (pSS) is a common autoimmune disease. Here, we performed the first proteome and phosphoproteome analyses of peripheral blood mononuclear cells in pSS patients to obtain a comprehensive profile and identify the potential crucial proteins and pathways for the screening and evaluation of pSS patients. Peripheral blood mononuclear cells from 8 pSS-confirmed patients (American-European Consensus Group Criteria, 2002) and 10 normal controls were selected. Label-free quantitative proteomics was utilized to obtain quantitative information. In total, 787 proteins were identified as differentially expressed proteins, and 175 phosphosites on 123 proteins were identified as differentially phosphorylated proteins. We performed functional enrichment analyses with these proteins and phosphoproteins based on public database. Furthermore, protein-protein interaction network analyses were performed by using multiple algorithms. Using module and hub protein analyses, we identified 16 modules for the proteins, 2 clusters for the phosphoproteins and selected the top 10 hub proteins. Finally, we identified 22 motifs using motif analysis of the phosphosites and found 17 newly identified motifs, while 6 motifs were experimentally verified for known protein kinases. The findings distinguished pSS patients from normal controls at the peripheral blood mononuclear cells level and revealed potential candidates for use in pSS diagnosis.
Collapse
Affiliation(s)
- Shaoying Huang
- Department of Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen People’s Hospital, The First Affiliated Hospital Southern University of Science and Technology, The Second Clinical Medical College of Jinan University, Shenzhen 518020, Guangdong, China
| | - Fengping Zheng
- Department of Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen People’s Hospital, The First Affiliated Hospital Southern University of Science and Technology, The Second Clinical Medical College of Jinan University, Shenzhen 518020, Guangdong, China
| | - Lixiong Liu
- Department of Rheumatology and Immunology, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen People’s Hospital, Shenzhen 518020, Guangdong, China
| | - Shuhui Meng
- Department of Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen People’s Hospital, The First Affiliated Hospital Southern University of Science and Technology, The Second Clinical Medical College of Jinan University, Shenzhen 518020, Guangdong, China
| | - Wanxia Cai
- Department of Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen People’s Hospital, The First Affiliated Hospital Southern University of Science and Technology, The Second Clinical Medical College of Jinan University, Shenzhen 518020, Guangdong, China
| | - Cantong Zhang
- Department of Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen People’s Hospital, The First Affiliated Hospital Southern University of Science and Technology, The Second Clinical Medical College of Jinan University, Shenzhen 518020, Guangdong, China
| | - Weier Dai
- College of Natural Science, University of Texas at Austin, Austin, TX 78712, USA
| | - Dongzhou Liu
- Department of Rheumatology and Immunology, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen People’s Hospital, Shenzhen 518020, Guangdong, China
| | - Xiaoping Hong
- Department of Rheumatology and Immunology, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen People’s Hospital, Shenzhen 518020, Guangdong, China
| | - Donge Tang
- Department of Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen People’s Hospital, The First Affiliated Hospital Southern University of Science and Technology, The Second Clinical Medical College of Jinan University, Shenzhen 518020, Guangdong, China
| | - Yong Dai
- Department of Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen People’s Hospital, The First Affiliated Hospital Southern University of Science and Technology, The Second Clinical Medical College of Jinan University, Shenzhen 518020, Guangdong, China.,Guangxi Key Laboratory of Metabolic Disease Research, Nephrology Department of Guilin, Guilin 541002, China
| |
Collapse
|
42
|
Pös Z, Pös O, Styk J, Mocova A, Strieskova L, Budis J, Kadasi L, Radvanszky J, Szemes T. Technical and Methodological Aspects of Cell-Free Nucleic Acids Analyzes. Int J Mol Sci 2020; 21:ijms21228634. [PMID: 33207777 PMCID: PMC7697251 DOI: 10.3390/ijms21228634] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 11/12/2020] [Accepted: 11/13/2020] [Indexed: 02/07/2023] Open
Abstract
Analyzes of cell-free nucleic acids (cfNAs) have shown huge potential in many biomedical applications, gradually entering several fields of research and everyday clinical care. Many biological properties of cfNAs can be informative to gain deeper insights into the function of the organism, such as their different types (DNA, RNAs) and subtypes (gDNA, mtDNA, bacterial DNA, miRNAs, etc.), forms (naked or vesicle bound NAs), fragmentation profiles, sequence composition, epigenetic modifications, and many others. On the other hand, the workflows of their analyzes comprise many important steps, from sample collection, storage and transportation, through extraction and laboratory analysis, up to bioinformatic analyzes and statistical evaluations, where each of these steps has the potential to affect the outcome and informational value of the performed analyzes. There are, however, no universal or standard protocols on how to exactly proceed when analyzing different cfNAs for different applications, at least according to our best knowledge. We decided therefore to prepare an overview of the available literature and products commercialized for cfNAs processing, in an attempt to summarize the benefits and limitations of the currently available approaches, devices, consumables, and protocols, together with various factors influencing the workflow, its processes, and outcomes.
Collapse
Affiliation(s)
- Zuzana Pös
- Institute of Clinical and Translational Research, Biomedical Research Center, Slovak Academy of Sciences, 845 05 Bratislava, Slovakia; (Z.P.); (A.M.); (L.K.)
- Department of Molecular Biology, Faculty of Natural Sciences, Comenius University, 841 04 Bratislava, Slovakia;
- Geneton Ltd., 841 04 Bratislava, Slovakia; (L.S.); (J.B.)
| | - Ondrej Pös
- Department of Molecular Biology, Faculty of Natural Sciences, Comenius University, 841 04 Bratislava, Slovakia;
- Geneton Ltd., 841 04 Bratislava, Slovakia; (L.S.); (J.B.)
- Comenius University Science Park, Comenius University, 841 04 Bratislava, Slovakia;
| | - Jakub Styk
- Comenius University Science Park, Comenius University, 841 04 Bratislava, Slovakia;
- Faculty of Medicine, Institute of Medical Biology, Genetics and Clinical Genetics, 811 08 Bratislava, Slovakia
| | - Angelika Mocova
- Institute of Clinical and Translational Research, Biomedical Research Center, Slovak Academy of Sciences, 845 05 Bratislava, Slovakia; (Z.P.); (A.M.); (L.K.)
- Department of Molecular Biology, Faculty of Natural Sciences, Comenius University, 841 04 Bratislava, Slovakia;
| | | | - Jaroslav Budis
- Geneton Ltd., 841 04 Bratislava, Slovakia; (L.S.); (J.B.)
- Comenius University Science Park, Comenius University, 841 04 Bratislava, Slovakia;
- Slovak Center of Scientific and Technical Information, 811 04 Bratislava, Slovakia
| | - Ludevit Kadasi
- Institute of Clinical and Translational Research, Biomedical Research Center, Slovak Academy of Sciences, 845 05 Bratislava, Slovakia; (Z.P.); (A.M.); (L.K.)
- Department of Molecular Biology, Faculty of Natural Sciences, Comenius University, 841 04 Bratislava, Slovakia;
| | - Jan Radvanszky
- Institute of Clinical and Translational Research, Biomedical Research Center, Slovak Academy of Sciences, 845 05 Bratislava, Slovakia; (Z.P.); (A.M.); (L.K.)
- Department of Molecular Biology, Faculty of Natural Sciences, Comenius University, 841 04 Bratislava, Slovakia;
- Comenius University Science Park, Comenius University, 841 04 Bratislava, Slovakia;
- Correspondence: (J.R.); (T.S.); Tel.: +421-2-60296637 (J.R.); +421-2-9026-8807 (T.S.)
| | - Tomas Szemes
- Department of Molecular Biology, Faculty of Natural Sciences, Comenius University, 841 04 Bratislava, Slovakia;
- Geneton Ltd., 841 04 Bratislava, Slovakia; (L.S.); (J.B.)
- Comenius University Science Park, Comenius University, 841 04 Bratislava, Slovakia;
- Correspondence: (J.R.); (T.S.); Tel.: +421-2-60296637 (J.R.); +421-2-9026-8807 (T.S.)
| |
Collapse
|
43
|
Jeon YS, Cha JK, Choi SH, Lee JH, Lee JS. Transcriptomic profiles and their correlations in saliva and gingival tissue biopsy samples from periodontitis and healthy patients. J Periodontal Implant Sci 2020; 50:313-326. [PMID: 33124209 PMCID: PMC7606893 DOI: 10.5051/jpis.1905460273] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 04/23/2020] [Accepted: 08/17/2020] [Indexed: 11/22/2022] Open
Abstract
Purpose This study was conducted to analyze specific RNA expression profiles in gingival tissue and saliva samples in periodontitis patients and healthy individuals, and to determine their correlations in light of the potential use of microarray-based analyses of saliva samples as a periodontal monitoring tool. Methods Gingival tissue biopsies and saliva samples from 22 patients (12 with severe periodontitis and 10 with a healthy periodontium) were analyzed using transcriptomic microarray analysis. Differential gene expression was assessed, and pathway and clustering analyses were conducted for the samples. The correlations between the results for the gingival tissue and saliva samples were analyzed at both the gene and pathway levels. Results There were 621 differentially expressed genes (DEGs; 320 upregulated and 301 downregulated) in the gingival tissue samples of the periodontitis group, and 154 DEGs (44 upregulated and 110 downregulated) in the saliva samples. Nine of these genes overlapped between the sample types. The periodontitis patients formed a distinct cluster group based on gene expression profiles for both the tissue and saliva samples. Database for Annotation, Visualization and Integrated Discovery analysis revealed 159 enriched pathways from the tissue samples of the periodontitis patients, as well as 110 enriched pathways In the saliva samples. Thirty-four pathways overlapped between the sample types. Conclusions The present results indicate the possibility of using the salivary transcriptome to distinguish periodontitis patients from healthy individuals. Further work is required to enhance the extraction of available RNA from saliva samples.
Collapse
Affiliation(s)
- Yoon Sun Jeon
- Department of Periodontology, Research Institute of Periodontal Regeneration, Yonsei University College of Dentistry, Seoul, Korea
| | - Jae Kook Cha
- Department of Periodontology, Research Institute of Periodontal Regeneration, Yonsei University College of Dentistry, Seoul, Korea
| | - Seong Ho Choi
- Department of Periodontology, Research Institute of Periodontal Regeneration, Yonsei University College of Dentistry, Seoul, Korea
| | - Ji Hyun Lee
- Department of Clinical Pharmacology and Therapeutics, Kyung Hee University College of Medicine, Seoul, Korea.,Department of Biomedical Science and Technology, Kyung Hee Medical Science Research Institute, Kyung Hee University, Seoul, Korea
| | - Jung Seok Lee
- Department of Periodontology, Research Institute of Periodontal Regeneration, Yonsei University College of Dentistry, Seoul, Korea.
| |
Collapse
|
44
|
Algohar A, Alqerban A. Levels of procalcitonin in saliva and peri-implant crevicular fluid in patients with peri-implant diseases and health. Arch Oral Biol 2020; 120:104931. [PMID: 33113457 DOI: 10.1016/j.archoralbio.2020.104931] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 09/14/2020] [Accepted: 09/14/2020] [Indexed: 02/08/2023]
Abstract
OBJECTIVE To evaluate the levels of procalcitonin in saliva and peri-implant crevicular fluid (PICF) among healthy and peri-implant disease patients and correlate these levels with clinical and radiographic peri-implant parameters. DESIGN Three groups of 20 participants each [Group-1: healthy, Group-2: peri-implant mucositis, and Group-3: peri-implantitis] were selected. Peri-implant plaque index, bleeding on probing, probing depth and crestal bone loss was assessed. PICF and saliva samples were evaluated for procalcitonin levels and analyzed using enzyme-linked immunosorbent assay. Kruskal-Wallis test was performed for comparisons among the study groups. Multiple comparisons were considered for Post hoc two-group comparisons using Bonferroni-corrections. The Spearman rank correlation coefficient analysis was used to analyze the correlation between procalcitonin levels of both fluids and clinical peri-implant parameters. RESULTS Group-3 demonstrated significantly higher values for peri-implant plaque index, bleeding on probing, probing depth, and crestal bone loss as compared to Group-1 and Group-2. Participants of both Group-2 and Group-3 reported significantly increased procalcitonin levels in saliva and PICF in comparison to Group-1. Significant positive correlations were found between PICF procalcitonin levels and bleeding on probing, probing depth, and crestal bone loss in Group-3 and significant positive correlation was found between PICF and bleeding on probing in Group-2. For salivary procalcitonin levels, a significant positive correlation was observed between procalcitonin and bleeding on probing in Group-3. CONCLUSIONS The outcome of this study suggests that procalcitonin might play a role in peri-implant inflammation, and higher procalcitonin levels is suggestive of a probable surrogate biomarker for peri-implant diseases.
Collapse
Affiliation(s)
- Ahmed Algohar
- Department of Prosthodontics, College of Dentistry, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia.
| | - Ali Alqerban
- Department of Preventive Dental Science, College of Dentistry, Dar Al Uloom University, Riyadh, Saudi Arabia; Department of Preventive Dental Science, College of Dentistry, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia.
| |
Collapse
|
45
|
Shodeinde AB, Murphy AC, Oldenkamp HF, Potdar AS, Ludolph CM, Peppas NA. Recent Advances in Smart Biomaterials for the Detection and Treatment of Autoimmune Diseases. ADVANCED FUNCTIONAL MATERIALS 2020; 30:1909556. [PMID: 33071713 PMCID: PMC7566744 DOI: 10.1002/adfm.201909556] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 01/15/2020] [Indexed: 05/07/2023]
Abstract
Autoimmune diseases are a group of debilitating illnesses that are often idiopathic in nature. The steady rise in the prevalence of these conditions warrants new approaches for diagnosis and treatment. Stimuli-responsive biomaterials also known as "smart", "intelligent" or "recognitive" biomaterials are widely studied for their applications in drug delivery, biosensing and tissue engineering due to their ability to produce thermal, optical, chemical, or structural changes upon interacting with the biological environment. This critical analysis highlights studies within the last decade that harness the recognitive capabilities of these biomaterials towards the development of novel detection and treatment options for autoimmune diseases.
Collapse
Affiliation(s)
- Aaliyah B. Shodeinde
- McKetta Department of Chemical Engineering, 200 E. Dean Keeton St. Stop C0400, Austin, TX, USA, 78712
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, 107 W Dean Keeton Street Stop C0800, Austin, TX, USA, 78712
| | - Andrew C. Murphy
- McKetta Department of Chemical Engineering, 200 E. Dean Keeton St. Stop C0400, Austin, TX, USA, 78712
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, 107 W Dean Keeton Street Stop C0800, Austin, TX, USA, 78712
| | - Heidi F. Oldenkamp
- McKetta Department of Chemical Engineering, 200 E. Dean Keeton St. Stop C0400, Austin, TX, USA, 78712
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, 107 W Dean Keeton Street Stop C0800, Austin, TX, USA, 78712
| | - Abhishek S. Potdar
- Department of Biomedical Engineering, The University of Texas at Austin, 107 W Dean Keeton Street Stop C0800, Austin, TX, USA, 78712
| | - Catherine M. Ludolph
- McKetta Department of Chemical Engineering, 200 E. Dean Keeton St. Stop C0400, Austin, TX, USA, 78712
| | - Nicholas A. Peppas
- McKetta Department of Chemical Engineering, 200 E. Dean Keeton St. Stop C0400, Austin, TX, USA, 78712
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, 107 W Dean Keeton Street Stop C0800, Austin, TX, USA, 78712
- Department of Biomedical Engineering, The University of Texas at Austin, 107 W Dean Keeton Street Stop C0800, Austin, TX, USA, 78712
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, 2409 University Ave. Stop A1900, Austin, TX, USA, 78712
- Department of Surgery and Perioperative Care, Dell Medical School, 1601 Trinity St., Bldg. B, Stop Z0800, Austin, TX, USA, 78712
- Department of Pediatrics, Dell Medical School, 1400 Barbara Jordan Blvd., Austin, TX, USA, 78723
| |
Collapse
|
46
|
Zgombić Popović V, Grgurević L, Trkulja V, Novak R, Negovetić-Vranić D. THE ROLE OF NEW TECHNOLOGIES IN DEFINING SALIVARY PROTEIN COMPOSITION FOLLOWING PLACEMENT OF FIXED ORTHODONTIC APPLIANCES - BREAKTHROUGH IN THE DEVELOPMENT OF NOVEL DIAGNOSTIC AND THERAPEUTIC PROCEDURES. Acta Clin Croat 2020; 59:480-488. [PMID: 34177058 PMCID: PMC8212653 DOI: 10.20471/acc.2020.59.03.12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Human saliva is rich in proteins of variable functions (e.g., enzymes, immunoglobulins, cytokines) and origin (blood plasma, salivary glands, or oral microflora). Circadian dynamics, volume and composition (electrolytes, pH, protein, etc.) of secreted saliva vary with local and systemic physiological and pathophysiological conditions. Therefore, the composition of saliva, protein in particular, has been intensively investigated to identify the potential markers and/or mechanisms of systemic and local diseases. Proteomic techniques used for the analysis of biological fluids have enabled great advances in salivary protein stabilization (as the main precondition for their analysis) and detection of those found in saliva in very low concentrations, including small proteins and peptides. This review brings the main characteristics of current proteomic techniques such as liquid chromatography-mass spectrometry, two-dimensional electrophoresis-mass spectrometry, and surface-enhanced laser desorption ionization/time of flight/mass spectrometry. These techniques enable simultaneous identification of hundreds and thousands of protein molecules, as well as identifying those of a potential biological value in particular states. This literature review is focused on the state-of-the-art and possibilities offered by proteomic techniques in analyzing the effects of orthodontic appliances on salivary protein composition and searching for potential markers of therapeutic success/failure or for the molecules by which therapeutic effects are achieved.
Collapse
Affiliation(s)
| | - Lovorka Grgurević
- 1Orthonova Dental Polyclinic, Zagreb, Croatia; 2Drago Perović Department of Anatomy, School of Medicine, University of Zagreb, Zagreb, Croatia; 3Department of Pharmacology, School of Medicine, University of Zagreb, Zagreb, Croatia; 4Center for Translational and Clinical Research, Department of Proteomics, School of Medicine, University of Zagreb, Zagreb, Croatia; 5Department of Pediatric and Preventive Dentistry, School of Dental Medicine, University of Zagreb, Zagreb, Croatia
| | - Vladimir Trkulja
- 1Orthonova Dental Polyclinic, Zagreb, Croatia; 2Drago Perović Department of Anatomy, School of Medicine, University of Zagreb, Zagreb, Croatia; 3Department of Pharmacology, School of Medicine, University of Zagreb, Zagreb, Croatia; 4Center for Translational and Clinical Research, Department of Proteomics, School of Medicine, University of Zagreb, Zagreb, Croatia; 5Department of Pediatric and Preventive Dentistry, School of Dental Medicine, University of Zagreb, Zagreb, Croatia
| | - Ruđer Novak
- 1Orthonova Dental Polyclinic, Zagreb, Croatia; 2Drago Perović Department of Anatomy, School of Medicine, University of Zagreb, Zagreb, Croatia; 3Department of Pharmacology, School of Medicine, University of Zagreb, Zagreb, Croatia; 4Center for Translational and Clinical Research, Department of Proteomics, School of Medicine, University of Zagreb, Zagreb, Croatia; 5Department of Pediatric and Preventive Dentistry, School of Dental Medicine, University of Zagreb, Zagreb, Croatia
| | - Dubravka Negovetić-Vranić
- 1Orthonova Dental Polyclinic, Zagreb, Croatia; 2Drago Perović Department of Anatomy, School of Medicine, University of Zagreb, Zagreb, Croatia; 3Department of Pharmacology, School of Medicine, University of Zagreb, Zagreb, Croatia; 4Center for Translational and Clinical Research, Department of Proteomics, School of Medicine, University of Zagreb, Zagreb, Croatia; 5Department of Pediatric and Preventive Dentistry, School of Dental Medicine, University of Zagreb, Zagreb, Croatia
| |
Collapse
|
47
|
Chen M, Luo R, Li S, Li H, Qin Y, Zhou D, Liu H, Gong X, Chang J. Paper-Based Strip for Ultrasensitive Detection of OSCC-Associated Salivary MicroRNA via CRISPR/Cas12a Coupling with IS-Primer Amplification Reaction. Anal Chem 2020; 92:13336-13342. [DOI: 10.1021/acs.analchem.0c02642] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Minghui Chen
- School of Life Sciences, Tianjin University and Tianjin Engineering Center of Micro-Nano Biomaterials and Detection-Treatment Technology (Tianjin), Tianjin 300072, China
| | - Ran Luo
- School of Life Sciences, Tianjin University and Tianjin Engineering Center of Micro-Nano Biomaterials and Detection-Treatment Technology (Tianjin), Tianjin 300072, China
| | - Shenghui Li
- Key Laboratory of Post-Neurotrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education and Tianjin, Tianjin Neurological Institute, Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Hengxuan Li
- School of Life Sciences, Tianjin University and Tianjin Engineering Center of Micro-Nano Biomaterials and Detection-Treatment Technology (Tianjin), Tianjin 300072, China
| | - Yi Qin
- School of Life Sciences, Tianjin University and Tianjin Engineering Center of Micro-Nano Biomaterials and Detection-Treatment Technology (Tianjin), Tianjin 300072, China
| | - Dianming Zhou
- Department of Toxicology, Tianjin Center for Disease Control and Prevention, Tianjin 300011, China
| | - Hao Liu
- Tianjin Stomatological Hospital, Tianjin 300041, China
| | - Xiaoqun Gong
- School of Life Sciences, Tianjin University and Tianjin Engineering Center of Micro-Nano Biomaterials and Detection-Treatment Technology (Tianjin), Tianjin 300072, China
| | - Jin Chang
- School of Life Sciences, Tianjin University and Tianjin Engineering Center of Micro-Nano Biomaterials and Detection-Treatment Technology (Tianjin), Tianjin 300072, China
| |
Collapse
|
48
|
Letawsky VH, Schreiber AM, Skoretz SA. A Tutorial on Saliva's Role in Swallowing With a Focus on Sjögren's Syndrome. AMERICAN JOURNAL OF SPEECH-LANGUAGE PATHOLOGY 2020; 29:1307-1319. [PMID: 32531172 DOI: 10.1044/2020_ajslp-19-00083] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Purpose Saliva is integral to swallowing and necessary for oral health. Understanding saliva's origin and properties is important for swallowing assessment and management. Diseases such as Sjögren's syndrome (SS) can affect saliva negatively, often contributing to dysphagia. Our objectives are to (a) highlight saliva's fundamental role in swallowing, (b) provide a bibliometric overview of literature pertaining to SS pathophysiology and effects on saliva, (c) explore implications of salivary changes on swallowing and quality of life in SS and other populations, and (d) provide suggestions for systematic saliva assessment in practice. Method This tutorial reviews saliva production, composition, and involvement in swallowing within health and disease. Using rapid review methodology, we outline the effect of SS on saliva and describe SS etiology, diagnosis, and treatment. We discuss formal saliva assessments and a multidisciplinary approach. Results Saliva plays a vital role in swallowing, particularly lubrication, bolus formation, and oral health. SS affects the salivary glands altering salivary flow rate and composition. We identified 55 studies (N) measuring salivary changes, grouping them according to four strata demarcated by SS classification criteria updates. For some, xerostomia, dysphagia, and reduced life quality result. Formal saliva assessments include the Clinical Oral Dryness Score, Xerostomia Inventory, and Secretion Rating Scale. Multidisciplinary care is optimal for patients with salivary changes. Conclusion Understanding salivary changes in disease may enhance understanding of swallowing and inform dysphagia practice. Expanding swallowing assessments with formal saliva evaluations, and patient perspectives thereof, may aid in developing bespoke treatments, ultimately improving outcomes and quality of life. Supplemental Material https://doi.org/10.23641/asha.12456449.
Collapse
Affiliation(s)
- Veronica H Letawsky
- School of Audiology and Speech Sciences, University of British Columbia, Vancouver, Canada
| | - Ann-Marie Schreiber
- School of Audiology and Speech Sciences, University of British Columbia, Vancouver, Canada
| | - Stacey A Skoretz
- School of Audiology and Speech Sciences, University of British Columbia, Vancouver, Canada
- Department of Critical Care Medicine, University of Alberta, Edmonton, Canada
- Centre for Heart Lung Innovation, St. Paul's Hospital, Providence Health Care, Vancouver, Canada
| |
Collapse
|
49
|
Yang Y, Ma L, Qiao X, Zhang X, Dong SF, Wu MT, Zhai K, Shi HZ. Salivary microRNAs show potential as biomarkers for early diagnosis of malignant pleural effusion. Transl Lung Cancer Res 2020; 9:1247-1257. [PMID: 32953502 PMCID: PMC7481620 DOI: 10.21037/tlcr-19-530] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background Malignant pleural effusion (MPE) is a common medical problem caused by multiple malignancies, especially lung cancers, and always comes along with a poor outcome. Early detection and diagnosis are important for improving the prognosis in patients with MPE. Salivary microRNAs (miRNAs) may represent a relatively convenient way for diagnosing MPE. We investigated the characteristics of salivary miRNAs of MPE patients, benign pleural effusion (BPE) patients, patients with a malignant tumor but without pleural effusion (MT), and healthy controls (HCs). We believe that they may show potential as a non-invasive and convenient biomarker for diagnosing MPE. Methods From January 1, 2019, to July 1, 2019, 57 MPE patients, 33 BPE patients, 50 MT patients, and 49 HCs were enrolled. To select candidate biomarkers, in the discovery phase, the salivary miRNA profiles were detected in three MPE patients and three HCs. Then, qPCR was used in the validation phase with 54 MPE patients, 33 BPE patients, 50 MT patients, and 46 HCs to assay the selected miRNAs. Results hsa-miR-4484 and hsa-miR-3663-3p were identified as potential biomarkers to diagnose MPE patients, with areas under the curve (AUC) of 0.768 and 0.666, respectively. The diagnostic efficacy was higher when the combination of both miRNAs was used, with an AUC of 0.802. No correlation was found between the volume of MPE and the expression of salivary miRNAs. Conclusions This study reports the characterization of salivary miRNAs collected from MPE patients. A combination of hsa-miR-4484 and hsa-miR-3663-3p showed potential discriminatory power for MPE detection, and it may be helpful for the early diagnosis of MPE, i.e., before the pleural effusion volume is too large.
Collapse
Affiliation(s)
- Yuan Yang
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Li Ma
- Department of Medical Oncology, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Xin Qiao
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Xin Zhang
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Shu-Feng Dong
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Min-Ting Wu
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Kan Zhai
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Huan-Zhong Shi
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
50
|
Aqrawi LA, Jensen JL, Fromreide S, Galtung HK, Skarstein K. Expression of NGAL-specific cells and mRNA levels correlate with inflammation in the salivary gland, and its overexpression in the saliva, of patients with primary Sjögren's syndrome. Autoimmunity 2020; 53:333-343. [PMID: 32686529 DOI: 10.1080/08916934.2020.1795140] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Salivary gland involvement is a characteristic feature of primary Sjögren's syndrome (pSS), where tissue destruction is mediated by infiltrating immune cells, and may be accompanied by the presence of adipose tissue. Optimally diagnosing this multifactorial disease requires the incorporation of additional routines. Screening for disease-specific biomarkers in biological fluid could be a promising approach to increase diagnostic accuracy. We have previously investigated disease biomarkers in saliva and tear fluid of pSS patients, identifying Neutrophil gelatinase-associated lipocalin (NGAL) as the most upregulated protein in pSS. In the current study, we aimed to explore for the first time NGAL expression at the site of inflammation in the pSS disease target organ. Immunohistochemical staining was conducted on minor salivary gland biopsies from 11 pSS patients and 11 non-SS sicca subjects, targeting NGAL-specific cells. Additional NGAL/PNAd double staining was performed to study NGAL expression in high endothelial venules, known as specialised vascular structures. Moreover, NGAL mRNA expression was measured utilising quantitative real-time polymerase chain reaction (qRT-PCR) on minor salivary gland biopsies from 15 pSS patients and 7 non-SS sicca individuals that served as tissue controls. Our results demonstrated NGAL expression in acinar and ductal epithelium within the salivary gland of pSS patients, where significantly greater levels of acinar NGAL were observed in pSS patients (p < .0018) when compared to non-SS subjects. Also, acinar expression positively correlated with focus score values (r 2 = 0.54, p < .02), while ductal epithelial expression showed a negative such correlation (r 2 = 0.74, p < .003). Some PNAD+ endothelial venules also expressed NGAL. An increase in NGAL staining with increased fatty replacement was also observed in pSS patients. Concurringly, a 27% increase in NGAL mRNA levels were also detected in the minor salivary glands of pSS patients when compared to non-SS tissue control subjects. In conclusion, there is a positive association between increase in NGAL expression and inflammation in the pSS disease target organ, which also coincides with its previously demonstrated upregulation in the saliva of pSS patients. Additional functional analyses are needed to better understand the immunological implications of this potential biomarker.
Collapse
Affiliation(s)
- Lara A Aqrawi
- Department of Oral Surgery and Oral Medicine, Institute of Clinical Odontology, University of Oslo, Oslo, Norway
| | - Janicke Liaaen Jensen
- Department of Oral Surgery and Oral Medicine, Institute of Clinical Odontology, University of Oslo, Oslo, Norway
| | - Siren Fromreide
- Gade Laboratory for Pathology, Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | | | - Kathrine Skarstein
- Gade Laboratory for Pathology, Department of Clinical Medicine, University of Bergen, Bergen, Norway.,Department of Pathology, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|