1
|
Yuan X, Wang J, Wang W, Song Y, Wu J, Du R. Microbiome alterations in primary Sjögren's syndrome: Regional dysbiosis and microbiome-targeted therapeutic strategies. Clin Immunol 2025; 273:110444. [PMID: 39947272 DOI: 10.1016/j.clim.2025.110444] [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: 12/11/2024] [Revised: 01/27/2025] [Accepted: 02/08/2025] [Indexed: 02/21/2025]
Abstract
Primary Sjögren's syndrome (pSS) is a complex autoimmune disease characterized by diverse clinical manifestations. While xerophthalmia and xerostomia are hallmark symptoms, the disease often involves multiple organ systems, including the kidneys, lungs, nervous system, and gastrointestinal tract, leading to systemic morbidity in severe cases. Despite extensive research, the precise pathogenesis of pSS remains unclear, likely involving infectious, hormonal, and genetic factors. Emerging evidence highlights the microbiome as a key contributor to autoimmune diseases, including pSS. Dysbiosis in the oral, ocular, gut, and genital microbiomes plays a critical role in disease onset, progression, and variability. This review summarizes current findings on microbiome alterations in pSS, emphasizing their role in pathogenesis and clinical features, and explores microbiome-targeted therapies. Understanding the role of the microbiome in pSS pathophysiology could advance disease management and inspire targeted therapeutic strategies.
Collapse
Affiliation(s)
- Xujing Yuan
- Department of Rheumatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Jun Wang
- Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Weiwei Wang
- Department of Rheumatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - You Song
- Department of Rheumatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Jiajia Wu
- Department of Rheumatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Rong Du
- Department of Rheumatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China.
| |
Collapse
|
2
|
Nagi R, Kumar SS, Sheth M, Deshpande A, Khan J. Association between oral microbiome dysbiosis and Sjogren Syndrome. A systematic review of clinical studies. Arch Oral Biol 2025; 172:106167. [PMID: 39798503 DOI: 10.1016/j.archoralbio.2024.106167] [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: 10/03/2024] [Revised: 12/17/2024] [Accepted: 12/19/2024] [Indexed: 01/15/2025]
Abstract
OBJECTIVES This systematic review investigates the association of oral microbiome dysbiosis with Sjogren Syndrome (SS). MATERIALS AND METHODS Indexed databases (PubMed/Medline, EMBASE, OVID, Web of Science, and Scopus) were independently searched for relevant manuscripts published until August 2024. Clinical studies on oral microbial flora count and diversity in SS patients were included. Risk of bias across individual studies was performed using the Risk of Bias in Nonrandomized Studies of Interventions tool. RESULTS Out of the initial 295 studies, 15 clinical studies met the selection criteria. The protocols were similar across the studies but varied in diagnostic criteria for SS, salivary flow estimation methods, dental and periodontal status findings, and the type of oral microbes observed. Out of 15 studies, 14 showed an alteration in the oral microbiome and differences in microbial diversity in SS patients. Higher oral microbial counts of Prevotella, Viellonella, and Firmicutes in SS were reported, whereas a higher prevalence of caries-associated bacteria Streptococcus, Lactobacillus, and Viellonella was found in SS patients. Overall, the studies had a low risk of bias. CONCLUSIONS The findings of the present review have shown the existence of significant oral microbial dysbiosis and differences in microbial diversity in SS patients compared to healthy subjects. Future well-designed longitudinal studies are needed to validate the results.
Collapse
Affiliation(s)
- Ravleen Nagi
- Department of Orofacial Pain and TMJ Disorders, Eastman Institute for Oral Health, University of Rochester, NY, USA.
| | - Sanjana Santhosh Kumar
- Department of General Dentistry, Eastman Institute for Oral Health, University of Rochester, NY, USA.
| | - Megha Sheth
- Department of General Dentistry, Eastman Institute for Oral Health, University of Rochester, NY, USA.
| | - Ashwini Deshpande
- SRM Dental College, Bharathi Salai, Ramapuram, Chennai, Tamil Nadu, India
| | - Junad Khan
- Department of Orofacial Pain and TMJ Disorders, Eastman Institute for Oral Health, University of Rochester, NY, USA.
| |
Collapse
|
3
|
Antman G, Ritzer L, Galor A, Verticchio Vercellin A, Siesky BA, Alabi D, Vayner J, Segev F, Harris A. The relationship between dry eye disease and human microbiota: A review of the science. Exp Eye Res 2024; 245:109951. [PMID: 38838972 PMCID: PMC11250917 DOI: 10.1016/j.exer.2024.109951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 05/15/2024] [Accepted: 06/02/2024] [Indexed: 06/07/2024]
Abstract
A complex relationship exists between human microbiota and the risk for ophthalmic disease. While the homeostatic composition of human microbiota is still being established, including what defines dysbiosis (i.e. changes in diversity and abundance), pilot research has begun to identify the potential influence of demographics, geography, and co-morbidities on the microbiota and describe their impact on ocular health. This review specifically focuses on the scientific relationships of the human oral and gut microbiota to dry eye disease (DED), a set of conditions impacting the tear film and ocular surface. Although data are sparse and often conflict across studies, the literature generally supports associations between microbial imbalance (dysbiosis) and DED and alterations in microbial diversity and abundance to specific aspects of DED. This review examines the relevant science and mechanistic relationships linking gut and oral dysbiosis and DED. Various physiochemical factors and therapeutic approaches that alter microbiota, including medications and fecal transplants are examined in relation to DED.
Collapse
Affiliation(s)
- Gal Antman
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai Hospital, New York, NY, USA; Department of Ophthalmology, Rabin Medical Center, Petach Tikwa, Israel; Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Lukas Ritzer
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai Hospital, New York, NY, USA
| | - Anat Galor
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami Veterans Affairs Medical Center, Miami, FL, USA
| | | | - Brent A Siesky
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai Hospital, New York, NY, USA
| | - Denise Alabi
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai Hospital, New York, NY, USA
| | - Jason Vayner
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai Hospital, New York, NY, USA
| | - Fani Segev
- Ophthalmology, Assuta Ashdod Medical Center, Goldman Medical School, Ben-Gurion University, Be'er Sheva, Israel
| | - Alon Harris
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai Hospital, New York, NY, USA.
| |
Collapse
|
4
|
Xiang X, Peng B, Liu K, Wang T, Ding P, Zhu Y, Cheng K, Ming Y. Prediction of delayed graft function by early salivary microbiota following kidney transplantation. Appl Microbiol Biotechnol 2024; 108:402. [PMID: 38951204 PMCID: PMC11217047 DOI: 10.1007/s00253-024-13236-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 06/11/2024] [Accepted: 06/14/2024] [Indexed: 07/03/2024]
Abstract
Delayed graft function (DGF) is a frequently observed complication following kidney transplantation (KT). Our prior research revealed dynamic shifts in salivary microbiota post-KT with immediate graft function (IGF), yet its behavior during DGF remains unexplored. Five recipients with DGF and 35 recipients with IGF were enrolled. Saliva samples were collected during the perioperative period, and 16S rRNA gene sequencing was performed. The salivary microbiota of IGFs changed significantly and gradually stabilized with the recovery of renal function. The salivary microbiota composition of DGFs was significantly different from that of IGFs, although the trend of variation appeared to be similar to that of IGFs. Salivary microbiota that differed significantly between patients with DGF and IGF at 1 day after transplantation were able to accurately distinguish the two groups in the randomForest algorithm (accuracy = 0.8333, sensitivity = 0.7778, specificity = 1, and area under curve = 0.85), with Selenomonas playing an important role. Bacteroidales (Spearman's r = - 0.4872 and p = 0.0293) and Veillonella (Spearmen's r = - 0.5474 and p = 0.0125) were significantly associated with the serum creatinine in DGF patients. Moreover, the significant differences in overall salivary microbiota structure between DGF and IGF patients disappeared upon long-term follow-up. This is the first study to investigate the dynamic changes in salivary microbiota in DGFs. Our findings suggested that salivary microbiota was able to predict DGF in the early stages after kidney transplantation, which might help the perioperative clinical management and early-stage intervention of kidney transplant recipients. KEY POINTS: • Salivary microbiota on the first day after KT could predict DGF. • Alterations in salivary taxa after KT are related to recovery of renal function.
Collapse
Affiliation(s)
- Xuyu Xiang
- The Transplantation Center of the Third Xiangya Hospital, Central South University, Changsha, 410013, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China
| | - Bo Peng
- The Transplantation Center of the Third Xiangya Hospital, Central South University, Changsha, 410013, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China
| | - Kai Liu
- The Transplantation Center of the Third Xiangya Hospital, Central South University, Changsha, 410013, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China
| | - Tianyin Wang
- The Transplantation Center of the Third Xiangya Hospital, Central South University, Changsha, 410013, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China
| | - Peng Ding
- The Transplantation Center of the Third Xiangya Hospital, Central South University, Changsha, 410013, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China
| | - Yi Zhu
- The Transplantation Center of the Third Xiangya Hospital, Central South University, Changsha, 410013, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China
| | - Ke Cheng
- The Transplantation Center of the Third Xiangya Hospital, Central South University, Changsha, 410013, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China
| | - Yingzi Ming
- The Transplantation Center of the Third Xiangya Hospital, Central South University, Changsha, 410013, China.
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China.
| |
Collapse
|
5
|
Shinde DB, Mahore JG, Giram PS, Singh SL, Sharda A, Choyan D, Musale S. Microbiota of Saliva: A Non-invasive Diagnostic Tool. Indian J Microbiol 2024; 64:328-342. [PMID: 39010986 PMCID: PMC11246313 DOI: 10.1007/s12088-024-01219-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 01/30/2024] [Indexed: 07/17/2024] Open
Abstract
Potential of salivary microbiota as a non-invasive diagnostic tool for various diseases are explained in the present review. Traditional diagnostic methods rely on blood, which has limitations in terms of collection and biomarker specificity. We discuss the concept of normal flora and how disruptions in oral microbiota can be indicative of diseases. Saliva, harboring a diverse microbial community, offers promise as a diagnostic biomarker source for oral and non-oral conditions. We delve into the role of microbial dysbiosis in disease pathogenesis and the prospects of using biological indicators like dysbiosis for diagnosis, prediction, and monitoring. This review also emphasizes the significance of saliva microbiota in advancing early disease detection and timely intervention. We addressed the following research question and objectives: Can the microbiota of saliva serve as a non-invasive diagnostic tool for the early detection and monitoring of both oral and non-oral diseases? To achieve this, we will explore the normal flora of microorganisms in the oral cavity, the impact of microbial dysbiosis, and the potential of using specific pathogenic microorganisms as biomarkers. Additionally, we will investigate the correlation between oral and non-oral diseases by analyzing total saliva or site-specific dental biofilms for signs of symbiosis or dysbiosis. This research seeks to contribute valuable insights into the development of a non-invasive diagnostic approach with broad applications in healthcare.
Collapse
Affiliation(s)
- Dasharath B Shinde
- Symbiosis School of Biological Sciences (SSBS), Symbiosis International (Deemed University), Pune, 412115 India
| | - Jayashri G Mahore
- Dr. D. Y. Patil Institute of Pharmaceutical Sciences and Research, Pimpri, Pune, 411018 India
- Sinhgad College of Pharmacy, Vadgaon (Bk.), Pune, 411041 India
| | - Prabhanjan S Giram
- Dr. D. Y. Patil Institute of Pharmaceutical Sciences and Research, Pimpri, Pune, 411018 India
- Department of Pharmaceutical Sciences, The State University of New York, Buffalo, NY 14214 USA
| | - Shaktikumar L Singh
- Dr. D. Y. Patil Institute of Pharmaceutical Sciences and Research, Pimpri, Pune, 411018 India
| | - Aditi Sharda
- Dr. D. Y. Patil Institute of Pharmaceutical Sciences and Research, Pimpri, Pune, 411018 India
| | - Divya Choyan
- Dr. D. Y. Patil Institute of Pharmaceutical Sciences and Research, Pimpri, Pune, 411018 India
| | - Shubham Musale
- Dr. D. Y. Patil Institute of Pharmaceutical Sciences and Research, Pimpri, Pune, 411018 India
| |
Collapse
|
6
|
Estaleen RA, Reilly CM, Luo XM. A double-edged sword: interactions of CX 3CL1/CX 3CR1 and gut microbiota in systemic lupus erythematosus. Front Immunol 2024; 14:1330500. [PMID: 38299151 PMCID: PMC10828040 DOI: 10.3389/fimmu.2023.1330500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 12/28/2023] [Indexed: 02/02/2024] Open
Abstract
Systemic lupus erythematosus (SLE) is a systemic chronic disease initiated by an abnormal immune response to self and can affect multiple organs. SLE is characterized by the production of autoantibodies and the deposition of immune complexes. In regard to the clinical observations assessed by rheumatologists, several chemokines and cytokines also contribute to disease progression. One such chemokine and adhesion molecule is CX3CL1 (otherwise known as fractalkine). CX3CL1 is involved in cell trafficking and inflammation through recognition by its receptor, CX3CR1. The CX3CL1 protein consists of a chemokine domain and a mucin-like stalk that allows it to function both as a chemoattractant and as an adhesion molecule. In inflammation and specifically lupus, the literature displays contradictory evidence for the functions of CX3CL1/CX3CR1 interactions. In addition, the gut microbiota has been shown to play an important role in the pathogenesis of SLE. This review highlights current studies that illustrate the interactions of the gut microbiota and CX3CR1 in SLE.
Collapse
Affiliation(s)
- Rana A. Estaleen
- Department of Biomedical Sciences and Pathobiology, Virgnia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| | - Christopher M. Reilly
- Biomedical Sciences, Edward Via College of Osteopathic Medicine, Blacksburg, VA, United States
| | - Xin M. Luo
- Department of Biomedical Sciences and Pathobiology, Virgnia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| |
Collapse
|
7
|
Amarnani A, Silverman GJ. Understanding the roles of the microbiome in autoimmune rheumatic diseases. RHEUMATOLOGY AND IMMUNOLOGY RESEARCH 2023; 4:177-187. [PMID: 38125641 PMCID: PMC10729600 DOI: 10.2478/rir-2023-0027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 08/06/2023] [Indexed: 12/23/2023]
Abstract
The gut microbiome represents a potential promising therapeutic target for autoimmune diseases. This review summarizes the current knowledge on the links between the gut microbiome and several autoimmune rheumatic diseases including rheumatoid arthritis (RA), systemic lupus erythematosus (SLE) spondyloarthropathies (SpA), Sjogren's syndrome (SS), and systemic sclerosis (SSc). Evidence from studies of RA and SLE patients suggests that alterations in the gut microbiome composition and function contribute to disease development and progression through increased gut permeability, with microbes and microbial metabolites driving an excessive systemic activation of the immune system. Also, there is growing evidence that gut dysbiosis and subsequent immune cell activation may contribute to disease pathogenesis in SpA and SS. For SSc, there are fewer, but these are still informative, reports on alterations in the gut microbiome. In general, the complex interplay between the microbiome and the immune system is still not fully understood. Here we discuss the current knowledge of the link between the gut microbiome and autoimmune rheumatic diseases, highlighting potentially fertile areas for future research and make considerations on the potential benefits of strategies that restore gut microbiome homeostasis.
Collapse
Affiliation(s)
- Abhimanyu Amarnani
- Department of Medicine, NYU Grossman School of Medicine, New York, NYUSA
| | - Gregg J. Silverman
- Department of Medicine, NYU Grossman School of Medicine, New York, NYUSA
| |
Collapse
|
8
|
He W, Lu Y, Shi R, An Q, Zhao J, Gao X, Zhang L, Ma D. Application of omics in Sjögren's syndrome. Inflamm Res 2023; 72:2089-2109. [PMID: 37878024 DOI: 10.1007/s00011-023-01797-x] [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: 08/07/2023] [Revised: 08/27/2023] [Accepted: 09/10/2023] [Indexed: 10/26/2023] Open
Abstract
OBJECTIVE The pathogenesis, diagnosis, and treatment of Sjögren's syndrome (SS) face many challenges, and there is an urgent need to develop new technologies to improve our understanding of SS. METHODS By searching the literature published domestically and internationally in the past 20 years, this artical reviewed the research of various omics techniques in SS. RESULTS Omics technology provided valuable insights into the pathogenesis, early diagnosis, condition and efficacy evaluation of SS. It is helpful to reveal the pathogenesis of the disease and explore new treatment schemes, which will open a new era for the study of SS. CONCLUSION At present, omics research has made some gratifying achievements, but there are still many uncertainties. Therefore, in the future, we should improve research techniques, standardize the collection of samples, and adopt a combination of multi-omics techniques to jointly study the pathogenesis of SS and provide new schemes for its treatment.
Collapse
Affiliation(s)
- Wenqin He
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
- Shanxi Province Clinical Research Center for Dermatologic and Immunologic Diseases (Rheumatic Diseases), Taiyuan, China
- Shanxi Province Clinical Theranostics Technology Innovation Center for Immunologic and Rheumatic Diseases, Taiyuan, China
| | - Yangyang Lu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
- Shanxi Province Clinical Research Center for Dermatologic and Immunologic Diseases (Rheumatic Diseases), Taiyuan, China
- Shanxi Province Clinical Theranostics Technology Innovation Center for Immunologic and Rheumatic Diseases, Taiyuan, China
| | - Rongjing Shi
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
- Shanxi Province Clinical Research Center for Dermatologic and Immunologic Diseases (Rheumatic Diseases), Taiyuan, China
- Shanxi Province Clinical Theranostics Technology Innovation Center for Immunologic and Rheumatic Diseases, Taiyuan, China
| | - Qi An
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
- Shanxi Province Clinical Research Center for Dermatologic and Immunologic Diseases (Rheumatic Diseases), Taiyuan, China
- Shanxi Province Clinical Theranostics Technology Innovation Center for Immunologic and Rheumatic Diseases, Taiyuan, China
| | - Jingwen Zhao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
- Shanxi Province Clinical Research Center for Dermatologic and Immunologic Diseases (Rheumatic Diseases), Taiyuan, China
- Shanxi Province Clinical Theranostics Technology Innovation Center for Immunologic and Rheumatic Diseases, Taiyuan, China
| | - Xinnan Gao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
- Shanxi Province Clinical Research Center for Dermatologic and Immunologic Diseases (Rheumatic Diseases), Taiyuan, China
- Shanxi Province Clinical Theranostics Technology Innovation Center for Immunologic and Rheumatic Diseases, Taiyuan, China
| | - Liyun Zhang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
- Shanxi Province Clinical Research Center for Dermatologic and Immunologic Diseases (Rheumatic Diseases), Taiyuan, China
- Shanxi Province Clinical Theranostics Technology Innovation Center for Immunologic and Rheumatic Diseases, Taiyuan, China
| | - Dan Ma
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China.
- Shanxi Province Clinical Research Center for Dermatologic and Immunologic Diseases (Rheumatic Diseases), Taiyuan, China.
- Shanxi Province Clinical Theranostics Technology Innovation Center for Immunologic and Rheumatic Diseases, Taiyuan, China.
- Shanxi Academy of Advanced Research and Innovation, Taiyuan, 030032, China.
| |
Collapse
|
9
|
Saúco C, Rus MJ, Nieto MR, Barros C, Cantiga-Silva C, Lendines-Cordero D, Calderer-Ortiz M, Zurita-García M, Arias-Herrera S, Monsalve-Guil L, Segura-Egea JJ, Simon-Soro A. Hyposalivation but not Sjögren's syndrome associated with microbial dysbiosis in women. Front Microbiol 2023; 14:1240891. [PMID: 37869670 PMCID: PMC10588445 DOI: 10.3389/fmicb.2023.1240891] [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: 06/15/2023] [Accepted: 09/18/2023] [Indexed: 10/24/2023] Open
Abstract
Background Saliva modulates the environment of the oral biofilm through pH buffer, microbial attachment to host surfaces, and nutritional source. The ecology of stress occurs when a physical factor adversely impacts an ecosystem or its biotic components. Therefore, reduced salivary flow can affect oral-host balance. The leading causes of hyposalivation include disease-associated Sjögren's syndrome (SS) and menopausal women as aging-associated. However, little is known about the oral microbiome integrated with sex hormones in hyposalivation. This study aimed to characterize the hyposalivation microbiome caused by aging or disease affecting the salivary glands in women. Methods We included 50 women older than 40 years of age in any menopausal phase. We collected stimulated saliva from 25 women diagnosed with SS (SS) and 25 without SS (non-SS). The bacterial profile of the patients was obtained by 16S rRNA sequencing. Bioinformatics analysis used machine learning to analyze the cohort's signs, symptoms, and bacterial profile. Salivary estradiol as a sex hormone variation level was determined. Results We obtained that 79% of the SS group, and 52% of the non-SS group had hyposalivation. We found a negatively correlated Prevotella-age and Rothia-estradiol in the SS group. Highlight, we found that the cause of the hyposalivation in the study did not explain differences in microbial diversity comparing non-SS and SS groups. Therefore, microbial communities found in hyposalivation but not related to systemic conditions suggest that changes in the oral environment might underpin host-microbial balance. Conclusion The salivary microbiome was similar in women with and without SS. However, hyposalivation showed two distinctive clusters associated with the bacterial population profiles. Our study suggests that local ecological disturbances could drive the change in the microbiome.
Collapse
Affiliation(s)
- Carlos Saúco
- Department of Stomatology, Faculty of Dentistry, University of Seville, Seville, Spain
| | - Maria J. Rus
- Department of Stomatology, Faculty of Dentistry, University of Seville, Seville, Spain
| | - María R. Nieto
- Department of Stomatology, Faculty of Dentistry, University of Seville, Seville, Spain
| | - Carolina Barros
- Department of Preventive and Restorative Dentistry, Dental School, São Paulo State University (UNESP), Araçatuba, Brazil
| | - Cristiane Cantiga-Silva
- Department of Preventive and Restorative Dentistry, Dental School, São Paulo State University (UNESP), Araçatuba, Brazil
| | | | - Marta Calderer-Ortiz
- Department of Stomatology, Faculty of Dentistry, University of Seville, Seville, Spain
| | - Miriam Zurita-García
- Department of Stomatology, Faculty of Dentistry, University of Seville, Seville, Spain
| | - Santiago Arias-Herrera
- Department of Dentistry, Faculty of Health Sciences, Universidad Europea de Valencia, Valencia, Spain
| | - Loreto Monsalve-Guil
- Department of Stomatology, Faculty of Dentistry, University of Seville, Seville, Spain
| | - Juan José Segura-Egea
- Department of Stomatology, Faculty of Dentistry, University of Seville, Seville, Spain
| | - Aurea Simon-Soro
- Department of Stomatology, Faculty of Dentistry, University of Seville, Seville, Spain
| |
Collapse
|
10
|
Bustos-Lobato L, Rus MJ, Saúco C, Simon-Soro A. Oral microbial biomap in the drought environment: Sjogren's syndrome. Mol Oral Microbiol 2023; 38:400-407. [PMID: 37767604 DOI: 10.1111/omi.12435] [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: 07/18/2023] [Revised: 09/11/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023]
Abstract
Sjogren's syndrome (SS) is an autoimmune disease that affects primarily the salivary glands, making perturbations in the oral ecosystem and potential factors of salivary flow that influence the onset and development of the disease. The oral cavity contains diverse microorganisms that inhabit various niches such as the oral microbial "biomap." It does not seem specific enough to establish a characteristic microbiome, given the diversity of clinical manifestations, variable rates of salivary secretion, and influential risk factors in patients with SS. This review discusses the biogeography of the oral microbiome in patients with SS such as saliva, tongue, tooth, mucosa, and gum. The microorganisms that were more abundant in the different oral niches were Gram-positive species, suggesting a higher survival of cell wall bacteria in this arid oral environment. Reduced salivary flow appears not to be linked to the cause of dysbiosis alone but influences host-associated risk factors. However, much work remains to be done to establish the role of the microbiome in the etiopathogenesis of autoimmune diseases such as SS. Future studies of the microbiome in autoimmunity will shed light on the role of specific microorganisms that have never been linked before with SS.
Collapse
Affiliation(s)
- Laura Bustos-Lobato
- Facultad de Odontología, Departamento de Estomatología, Universidad de Sevilla, Sevilla, Spain
| | - Maria J Rus
- Facultad de Odontología, Departamento de Estomatología, Universidad de Sevilla, Sevilla, Spain
| | - Carlos Saúco
- Facultad de Odontología, Departamento de Estomatología, Universidad de Sevilla, Sevilla, Spain
| | - Aurea Simon-Soro
- Facultad de Odontología, Departamento de Estomatología, Universidad de Sevilla, Sevilla, Spain
| |
Collapse
|
11
|
Cao Y, Lu H, Xu W, Zhong M. Gut microbiota and Sjögren's syndrome: a two-sample Mendelian randomization study. Front Immunol 2023; 14:1187906. [PMID: 37383227 PMCID: PMC10299808 DOI: 10.3389/fimmu.2023.1187906] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 06/02/2023] [Indexed: 06/30/2023] Open
Abstract
Background The link between the gut microbiota (GM) and Sjögren's Syndrome (SS) is well-established and apparent. Whether GM is causally associated with SS is uncertain. Methods The MiBioGen consortium's biggest available genome-wide association study (GWAS) meta-analysis (n=13,266) was used as the basis for a two-sample Mendelian randomization study (TSMR). The causal relationship between GM and SS was investigated using the inverse variance weighted, MR-Egger, weighted median, weighted model, MR-PRESSO, and simple model methods. In order to measure the heterogeneity of instrumental variables (IVs), Cochran's Q statistics were utilized. Results The results showed that genus Fusicatenibacter (odds ratio (OR) = 1.418, 95% confidence interval (CI), 1.072-1.874, P = 0.0143) and genus Ruminiclostridium9 (OR = 1.677, 95% CI, 1.050-2.678, P = 0.0306) were positively correlated with the risk of SS and family Porphyromonadaceae (OR = 0.651, 95% CI, 0.427-0.994, P = 0.0466), genus Subdoligranulum (OR = 0.685, 95% CI, 0.497-0.945, P = 0.0211), genus Butyricicoccus (OR = 0.674, 95% CI, 0.470-0.967, P = 0.0319) and genus Lachnospiraceae (OR = 0.750, 95% CI, 0.585-0.961, P = 0.0229) were negatively correlated with SS risk using the inverse variance weighted (IVW) technique. Furthermore, four GM related genes: ARAP3, NMUR1, TEC and SIRPD were significant causally with SS after FDR correction (FDR<0.05). Conclusions This study provides evidence for either positive or negative causal effects of GM composition and its related genes on SS risk. We want to provide novel approaches for continued GM and SS-related research and therapy by elucidating the genetic relationship between GM and SS.
Collapse
Affiliation(s)
- Yu Cao
- School of Medicine, Xiamen University, Xiamen, China
| | - Hao Lu
- School of Medicine, Xiamen University, Xiamen, China
| | - Wangzi Xu
- School of Medicine, Xiamen University, Xiamen, China
| | - Ming Zhong
- Department of Oral Histopathology, School and Hospital of Stomatology, China Medical University, Liaoning Province Key Laboratory of Oral Disease, Shenyang, Liaoning, China
| |
Collapse
|
12
|
Kozhakhmetov S, Babenko D, Issilbayeva A, Nurgaziyev M, Kozhakhmetova S, Meiramova A, Akhmetova Z, Kunz J, Ainabekova B, Marotta F, Kushugulova A. Oral Microbial Signature of Rheumatoid Arthritis in Female Patients. J Clin Med 2023; 12:3694. [PMID: 37297889 PMCID: PMC10253734 DOI: 10.3390/jcm12113694] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/18/2023] [Accepted: 05/22/2023] [Indexed: 06/12/2023] Open
Abstract
This study aimed to identify the oral microbial signature of Kazakh female rheumatoid arthritis (RA) patients. A total of 75 female patients who met the American College of Rheumatology 2010 classification criteria for RA and 114 healthy volunteers were included in the study. Amplicons of the 16S rRNA gene were sequenced to analyze the microbial composition. We identified significant differences in bacterial diversity and abundance between the RA and control groups, as measured by Shannon (p value = 0.0205) and Simpson (p value = 0.00152) indices. The oral samples from RA patients had higher bacterial diversity than those from non-RA volunteers. The RA samples had a higher relative abundance of Prevotellaceae and Leptotrichiaceae, but a lower content of butyrate and propionate-producing bacteria compared to the control group. The samples from patients in remission had a higher abundance of Treponema sp. and Absconditabacteriales (SR1), whereas those with low disease activity had higher levels of Porphyromonas and those with high RA activity had higher levels of Staphylococcus. A positive correlation was found between the taxa Prevotella_9 and serum levels of antibodies to cyclic citrullinated peptide (ACPA) and rheumatoid factor (RF). The predicted functional pattern of the ACPA+/RF- and ACPA+/RF+ seropositive groups was characterized by increased ascorbate metabolism, degradation of glycosaminoglycans, and reduced biodegradation of xenobiotics. These findings suggest that the functional pattern of the microflora should be considered when selecting a therapeutic strategy for RA in order to provide a personalized approach.
Collapse
Affiliation(s)
- Samat Kozhakhmetov
- Laboratory of Microbiome, Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Astana Z05H0P9, Kazakhstan
| | | | - Argul Issilbayeva
- Laboratory of Microbiome, Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Astana Z05H0P9, Kazakhstan
- Department of Internal Medicine with the Course of Gastroenterology, Endocrinology and Pulmonology, NJSC Astana Medical University, Astana 010000, Kazakhstan
| | - Madiyar Nurgaziyev
- Laboratory of Microbiome, Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Astana Z05H0P9, Kazakhstan
| | | | - Assel Meiramova
- Laboratory of Microbiome, Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Astana Z05H0P9, Kazakhstan
- Department of Internal Medicine with the Course of Gastroenterology, Endocrinology and Pulmonology, NJSC Astana Medical University, Astana 010000, Kazakhstan
| | - Zhanar Akhmetova
- Laboratory of Microbiome, Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Astana Z05H0P9, Kazakhstan
- Department of Internal Medicine with the Course of Gastroenterology, Endocrinology and Pulmonology, NJSC Astana Medical University, Astana 010000, Kazakhstan
| | - Jeanette Kunz
- Department of Medicine, Nazarbayev University School of Medicine, Astana Z05H0P9, Kazakhstan
| | - Bayan Ainabekova
- Department of Internal Medicine with the Course of Gastroenterology, Endocrinology and Pulmonology, NJSC Astana Medical University, Astana 010000, Kazakhstan
| | - Francesco Marotta
- ReGenera R&D International for Aging Intervention, 20144 Milan, Italy
| | - Almagul Kushugulova
- Laboratory of Microbiome, Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Astana Z05H0P9, Kazakhstan
| |
Collapse
|
13
|
Huang X, Huang X, Huang Y, Zheng J, Lu Y, Mai Z, Zhao X, Cui L, Huang S. The oral microbiome in autoimmune diseases: friend or foe? J Transl Med 2023; 21:211. [PMID: 36949458 PMCID: PMC10031900 DOI: 10.1186/s12967-023-03995-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 02/15/2023] [Indexed: 03/24/2023] Open
Abstract
The human body is colonized by abundant and diverse microorganisms, collectively known as the microbiome. The oral cavity has more than 700 species of bacteria and consists of unique microbiome niches on mucosal surfaces, on tooth hard tissue, and in saliva. The homeostatic balance between the oral microbiota and the immune system plays an indispensable role in maintaining the well-being and health status of the human host. Growing evidence has demonstrated that oral microbiota dysbiosis is actively involved in regulating the initiation and progression of an array of autoimmune diseases.Oral microbiota dysbiosis is driven by multiple factors, such as host genetic factors, dietary habits, stress, smoking, administration of antibiotics, tissue injury and infection. The dysregulation in the oral microbiome plays a crucial role in triggering and promoting autoimmune diseases via several mechanisms, including microbial translocation, molecular mimicry, autoantigen overproduction, and amplification of autoimmune responses by cytokines. Good oral hygiene behaviors, low carbohydrate diets, healthy lifestyles, usage of prebiotics, probiotics or synbiotics, oral microbiota transplantation and nanomedicine-based therapeutics are promising avenues for maintaining a balanced oral microbiome and treating oral microbiota-mediated autoimmune diseases. Thus, a comprehensive understanding of the relationship between oral microbiota dysbiosis and autoimmune diseases is critical for providing novel insights into the development of oral microbiota-based therapeutic approaches for combating these refractory diseases.
Collapse
Affiliation(s)
- Xiaoyan Huang
- Department of Preventive Dentistry, Stomatological Hospital, School of Stomatology, Southern Medical University, Haizhu District, No.366 Jiangnan Da Dao Nan, Guangzhou, 510280, China
| | - Xiangyu Huang
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Haizhu District, No.366 Jiangnan Da Dao Nan, Guangzhou, 510280, China
| | - Yi Huang
- Department of Preventive Dentistry, Stomatological Hospital, School of Stomatology, Southern Medical University, Haizhu District, No.366 Jiangnan Da Dao Nan, Guangzhou, 510280, China
| | - Jiarong Zheng
- Department of Dentistry, The First Affiliated Hospital, Sun Yat-Sen University, Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Ye Lu
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Haizhu District, Guangzhou, 510280, China
| | - Zizhao Mai
- Department of Dentistry, The First Affiliated Hospital, Sun Yat-Sen University, Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Xinyuan Zhao
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Haizhu District, No.366 Jiangnan Da Dao Nan, Guangzhou, 510280, China.
| | - Li Cui
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Haizhu District, Guangzhou, 510280, China.
- Division of Oral Biology and Medicine, School of Dentistry, University of California, Los Angeles, CA, 90095, USA.
| | - Shaohong Huang
- Department of Preventive Dentistry, Stomatological Hospital, School of Stomatology, Southern Medical University, Haizhu District, No.366 Jiangnan Da Dao Nan, Guangzhou, 510280, China.
| |
Collapse
|
14
|
Wiriyakijja P, Niklander S, Santos-Silva AR, Shorrer MK, Simms ML, Villa A, Sankar V, Kerr AR, Riordain RN, Jensen SB, Delli K. World Workshop on Oral Medicine VIII: Development of a Core Outcome Set for Dry Mouth: A Systematic Review of Outcome Domains for Xerostomia. Oral Surg Oral Med Oral Pathol Oral Radiol 2023:S2212-4403(23)00068-8. [PMID: 37198047 DOI: 10.1016/j.oooo.2023.01.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/11/2022] [Accepted: 01/25/2023] [Indexed: 03/08/2023]
Abstract
OBJECTIVE The purpose of this study was to identify all outcome domains used in clinical studies of xerostomia, that is, subjective sensation of dry mouth. This study is part of the extended project "World Workshop on Oral Medicine Outcomes Initiative for the Direction of Research" to develop a core outcome set for dry mouth. STUDY DESIGN A systematic review was performed on MEDLINE, EMBASE, CINAHL, and Cochrane Central Register of Controlled Trials databases. All clinical and observational studies that assessed xerostomia in human participants from 2001 to 2021 were included. Information on outcome domains was extracted and mapped to the Core Outcome Measures in Effectiveness Trials taxonomy. Corresponding outcome measures were summarized. RESULTS From a total of 34,922 records retrieved, 688 articles involving 122,151 persons with xerostomia were included. There were 16 unique outcome domains and 166 outcome measures extracted. None of these domains or measures were consistently used across all the studies. The severity of xerostomia and physical functioning were the 2 most frequently assessed domains. CONCLUSION There is considerable heterogeneity in outcome domains and measures reported in clinical studies of xerostomia. This highlights the need for harmonization of dry mouth assessment to enhance comparability across studies and facilitate the synthesis of robust evidence for managing patients with xerostomia.
Collapse
|
15
|
High-Throughput Sequencing of Oral Microbiota in Candida Carriage Sjögren's Syndrome Patients: A Pilot Cross-Sectional Study. J Clin Med 2023; 12:jcm12041559. [PMID: 36836095 PMCID: PMC9964208 DOI: 10.3390/jcm12041559] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 02/13/2023] [Accepted: 02/14/2023] [Indexed: 02/18/2023] Open
Abstract
BACKGROUND This study sought to characterize the saliva microbiota of Candida carriage Sjögren's syndrome (SS) patients compared to oral candidiasis and healthy patients by high-throughput sequencing. METHODS Fifteen patients were included, with five Candida carriage SS patients (decayed, missing, and filled teeth (DMFT) score 22), five oral candidiasis patients (DMFT score 17), and five caries active healthy patients (DMFT score 14). Bacterial 16S rRNA was extracted from rinsed whole saliva. PCR amplification generated DNA amplicons of the V3-V4 hypervariable region, which were sequenced on an Illumina HiSeq 2500 sequencing platform and compared and aligned to the SILVA database. Taxonomy abundance and community structure diversity was analyzed using Mothur software v1.40.0. RESULTS A total of 1016/1298/1085 operational taxonomic units (OTUs) were obtained from SS patients/oral candidiasis patient/healthy patients. Treponema, Lactobacillus, Streptococcus, Selenomonas, and Veillonella were the primary genera in the three groups. The most abundant significantly mutative taxonomy (OTU001) was Veillonella parvula. Microbial diversity (alpha diversity and beta diversity) was significantly increased in SS patients. ANOSIM analyses revealed significantly different microbial compositional heterogeneity in SS patients compared to oral candidiasis and healthy patients. CONCLUSION Microbial dysbiosis differs significantly in SS patients independent of oral Candida carriage and DMFT.
Collapse
|
16
|
A clinically validated human saliva metatranscriptomic test for global systems biology studies. Biotechniques 2023; 74:31-44. [PMID: 36622006 DOI: 10.2144/btn-2022-0104] [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] [Indexed: 01/10/2023] Open
Abstract
The authors report here the development of a high-throughput, automated, inexpensive and clinically validated saliva metatranscriptome test that requires less than 100 μl of saliva. RNA is preserved at the time of sample collection, allowing for ambient-temperature transportation and storage for up to 28 days. Critically, the RNA preservative is also able to inactivate pathogenic microorganisms, rendering the samples noninfectious and allowing for safe and easy shipping. Given the unique set of convenience, low cost, safety and technical performance, this saliva metatranscriptomic test can be integrated into longitudinal, global-scale systems biology studies that will lead to an accelerated development of precision medicine, diagnostic and therapeutic tools.
Collapse
|
17
|
Georges FM, Do NT, Seleem D. Oral dysbiosis and systemic diseases. FRONTIERS IN DENTAL MEDICINE 2022. [DOI: 10.3389/fdmed.2022.995423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The aim of this mini review is to investigate the connection between oral microbiome dysbiosis and systemic diseases. Many systemic conditions can have oral manifestations and cause worsening in oral diseases. For example, uncontrolled type 2 diabetes has been associated with worsening of periodontal disease. Other inflammatory diseases or autoimmune diseases may predispose to oral mucositis, mucosal ulcers, xerostomia, and higher susceptibility to oral infections. This review will outline common systemic diseases, such as metabolic, cardiovascular, and immunologic disorders as they relate to oral manifestations and changes in the oral microbiome composition.
Collapse
|
18
|
Huang X, Chen X, Gong X, Xu Y, Xu Z, Gao X. Characteristics of salivary microbiota in children with obstructive sleep apnea: A prospective study with polysomnography. Front Cell Infect Microbiol 2022; 12:945284. [PMID: 36105146 PMCID: PMC9465092 DOI: 10.3389/fcimb.2022.945284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
ObjectivesThe present study aimed to investigate the characteristics of salivary microbiota of children with obstructive sleep apnea (OSA) and to assess longitudinal alterations in salivary microbiota before and after adenotonsillectomy.MethodsA set of cross-sectional samples consisted of 36 OSA children (17 boys and 19 girls, 7.47 ± 2.24 years old) and 22 controls (9 boys and 13 girls, 7.55 ± 2.48 years old) were included in the study, among which eight OSA children (five boys and three girls, 8.8 ± 2.0 years old) who underwent treatment of adenotonsillectomy were followed up after 1 year. Saliva samples were collected, and microbial profiles were analyzed by bioinformatics analysis based on 16S rRNA sequencing.ResultsIn cross-sectional samples, the OSA group had higher α-diversity as estimated by Chao1, Shannon, Simpson, Pielou_e, and observed species as compared with the control group (p < 0.05). β-Diversity based on the Bray–Curtis dissimilarities (p = 0.004) and Jaccard distances (p = 0.001) revealed a significant separation between the OSA group and control group. Nested cross-validated random forest classifier identified the 10 most important genera (Lactobacillus, Escherichia, Bifidobacterium, Capnocytophaga, Bacteroidetes_[G-7], Parvimonas, Bacteroides, Klebsiella, Lautropia, and Prevotella) that could differentiate OSA children from controls with an area under the curve (AUC) of 0.94. Linear discriminant analysis effect size (LEfSe) analysis revealed a significantly higher abundance of genera such as Prevotella (p = 0.027), Actinomyces (p = 0.015), Bifidobacterium (p < 0.001), Escherichia (p < 0.001), and Lactobacillus (p < 0.001) in the OSA group, among which Prevotella was further corroborated in longitudinal samples. Prevotella sp_HMT_396 was found to be significantly enriched in the OSA group (p = 0.02) with significantly higher levels as OSA severity increased (p = 0.014), and it had a lower abundance in the post-treatment group (p = 0.003) with a decline in each OSA child 1 year after adenotonsillectomy.ConclusionsA significantly higher microbial diversity and a significant difference in microbial composition and abundance were identified in salivary microbiota of OSA children compared with controls. Meanwhile, some characteristic genera (Prevotella, Actinomyces, Lactobacillus, Escherichia, and Bifidobacterium) were found in OSA children, among which the relationship between Prevotella spp. and OSA is worth further studies.
Collapse
Affiliation(s)
- Xin Huang
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
| | - Xuehui Chen
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
| | - Xu Gong
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
| | - Ying Xu
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
| | - Zhifei Xu
- Department of Respiratory Medicine, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
- *Correspondence: Xuemei Gao,
| | - Xuemei Gao
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
- *Correspondence: Xuemei Gao,
| |
Collapse
|
19
|
Chattopadhyay I, Lu W, Manikam R, Malarvili MB, Ambati RR, Gundamaraju R. Can metagenomics unravel the impact of oral bacteriome in human diseases? Biotechnol Genet Eng Rev 2022; 39:85-117. [PMID: 35861776 DOI: 10.1080/02648725.2022.2102877] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Oral microbial ecosystems are vital in maintaining the health of the oral cavity and the entire body. Oral microbiota is associated with the progression of oral diseases such as dental caries, periodontal diseases, head and neck cancer, and several systemic diseases such as cardiovascular disease, rheumatoid arthritis, adverse pregnancy outcomes, diabetes, lung infection, colorectal cancer, and pancreatic cancer. Buccal mucosa, tongue dorsum, hard palate, saliva, palatine tonsils, throat, keratinized gingiva, supra-gingival plaque, subgingival plaque, dentures, and lips are microbial habitats of the oral cavity. Porphyromonas gingivalis may have a role in the development of periodontal diseases, oral cancer, diabetes, and atherosclerotic disease. Fusobacterium nucleatum showed a higher abundance in periodontal diseases, oral and colon cancer, adverse pregnancy outcomes, diabetes, and rheumatoid arthritis. The higher abundance of Prevotella intermedia is typical in periodontal diseases, rheumatoid arthritis, and adverse pregnancy outcome. S. salivarius displayed higher abundance in both dental caries and OSCC. Oral bacteria may influence systemic diseases through inflammation by releasing pro inflammatory cytokines. Identification of oral bacteria using culture-dependent approaches and next-generation sequencing-based metagenomic approaches is believed to significantly identify the therapeutic targets and non-invasive diagnostic indicators in different human diseases. Oral bacteria in saliva could be exploited as a non-invasive diagnostic indicator for the early detection of oral and systemic disorders. Other therapeutic approaches such as the use of probiotics, green tea polyphenol, cold atmospheric plasma (CAP) therapy, antimicrobial photodynamic therapy, and antimicrobial peptides are used to inhibit the growth of biofilm formation by oral bacteria.
Collapse
Affiliation(s)
| | - Wenying Lu
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, University of Tasmania, Launceston, Tasmania, Australia
| | - Rishya Manikam
- Trauma and Emergency, University of Malaya, Kuala Lumpur, Malaysia
| | - M B Malarvili
- School of Biomedical and Health Science, Universiti Teknologi Malaysia, Skudai, Johor Bahru, Malaysia
| | - Ranga Rao Ambati
- Department of Biotechnology, Vignan`s Foundation for Science, Technology and Research (Deemed to be University), Guntur, Andhra Pradesh, India
| | - Rohit Gundamaraju
- ER stress and Mucosal immunology lab, School of Health Sciences, University of Tasmania, Launceston, Tasmania, Australia
| |
Collapse
|
20
|
Deng C, Xiao Q, Fei Y. A Glimpse Into the Microbiome of Sjögren’s Syndrome. Front Immunol 2022; 13:918619. [PMID: 35911741 PMCID: PMC9329934 DOI: 10.3389/fimmu.2022.918619] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 06/20/2022] [Indexed: 11/13/2022] Open
Abstract
Sjögren’s syndrome (SS) is a common chronic systemic autoimmune disease and its main characteristic is lymphoid infiltration of the exocrine glands, particularly the salivary and lacrimal glands, leading to sicca symptoms of the mouth and eyes. Growing evidence has shown that SS is also characterized by microbial perturbations like other autoimmune diseases. Significant alterations in diversity, composition, and function of the microbiota were observed in SS. The dysbiosis of the microbiome correlates with worse symptoms and higher disease severity, suggesting that dysbiosis may be of great importance in the pathogenesis of SS. In this review, we provide a general view of recent studies describing the microbiota alterations of SS, the possible pathways that may cause microbiota dysbiosis to trigger SS, and the existence of the gut-ocular/gut-oral axis in SS.
Collapse
|
21
|
van der Meulen TA, Vissink A, Bootsma H, Spijkervet FKL, Kroese FGM. Microbiome in Sjögren's syndrome: here we are. Ann Rheum Dis 2022; 81:e114. [PMID: 32699041 DOI: 10.1136/annrheumdis-2020-218213] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 06/10/2020] [Indexed: 11/04/2022]
Affiliation(s)
- Taco A van der Meulen
- Oral and Maxillofacial Surgery, Universitair Medisch Centrum Groningen, Groningen, The Netherlands
| | - Arjan Vissink
- Oral and Maxillofacial Surgery, Universitair Medisch Centrum Groningen, Groningen, The Netherlands
| | - Hendrika Bootsma
- Rheumatology and Clinical Immunology, University Medical Centre Groningen, Groningen, The Netherlands
| | - Fred K L Spijkervet
- Oral and Maxillofacial Surgery, Universitair Medisch Centrum Groningen, Groningen, The Netherlands
| | - Frans G M Kroese
- Rheumatology and Clinical Immunology, University Medical Centre Groningen, Groningen, The Netherlands
| |
Collapse
|
22
|
Manasson J, Blank RB, Scher JU. Response to: 'Microbiome in Sjögren's syndrome: here we are' by van der Meulen et al. Ann Rheum Dis 2022; 81:e115. [PMID: 32699036 DOI: 10.1136/annrheumdis-2020-218327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 06/25/2020] [Indexed: 11/03/2022]
Affiliation(s)
- Julia Manasson
- Division of Rheumatology, Department of Medicine, New York University Grossman School of Medicine, New York, New York, USA
| | - Rebecca B Blank
- Division of Rheumatology, Department of Medicine, New York University Grossman School of Medicine, New York, New York, USA
| | - Jose U Scher
- Division of Rheumatology, Department of Medicine, New York University Grossman School of Medicine, New York, New York, USA
| |
Collapse
|
23
|
Gao L, Cheng Z, Zhu F, Bi C, Shi Q, Chen X. The Oral Microbiome and Its Role in Systemic Autoimmune Diseases: A Systematic Review of Big Data Analysis. Front Big Data 2022; 5:927520. [PMID: 35844967 PMCID: PMC9277227 DOI: 10.3389/fdata.2022.927520] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 05/23/2022] [Indexed: 12/30/2022] Open
Abstract
Introduction Despite decades of research, systemic autoimmune diseases (SADs) continue to be a major global health concern and the etiology of these diseases is still not clear. To date, with the development of high-throughput techniques, increasing evidence indicated a key role of oral microbiome in the pathogenesis of SADs, and the alterations of oral microbiome may contribute to the disease emergence or evolution. This review is to present the latest knowledge on the relationship between the oral microbiome and SADs, focusing on the multiomics data generated from a large set of samples. Methodology By searching the PubMed and Embase databases, studies that investigated the oral microbiome of SADs, including systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), and Sjögren's syndrome (SS), were systematically reviewed according to the PRISMA guidelines. Results One thousand and thirty-eight studies were found, and 25 studies were included: three referred to SLE, 12 referred to RA, nine referred to SS, and one to both SLE and SS. The 16S rRNA sequencing was the most frequent technique used. HOMD was the most common database aligned to and QIIME was the most popular pipeline for downstream analysis. Alterations in bacterial composition and population have been found in the oral samples of patients with SAD compared with the healthy controls. Results regarding candidate pathogens were not always in accordance, but Selenomonas and Veillonella were found significantly increased in three SADs, and Streptococcus was significantly decreased in the SADs compared with controls. Conclusion A large amount of sequencing data was collected from patients with SAD and controls in this systematic review. Oral microbial dysbiosis had been identified in these SADs, although the dysbiosis features were different among studies. There was a lack of standardized study methodology for each study from the inclusion criteria, sample type, sequencing platform, and referred database to downstream analysis pipeline and cutoff. Besides the genomics, transcriptomics, proteomics, and metabolomics technology should be used to investigate the oral microbiome of patients with SADs and also the at-risk individuals of disease development, which may provide us with a better understanding of the etiology of SADs and promote the development of the novel therapies.
Collapse
Affiliation(s)
- Lu Gao
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Oral Diseases, Hangzhou, China
- Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Zijian Cheng
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Oral Diseases, Hangzhou, China
- Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Fudong Zhu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Oral Diseases, Hangzhou, China
- Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Chunsheng Bi
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Oral Diseases, Hangzhou, China
- Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Qiongling Shi
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Oral Diseases, Hangzhou, China
- Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Xiaoyan Chen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Oral Diseases, Hangzhou, China
- Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
- *Correspondence: Xiaoyan Chen
| |
Collapse
|
24
|
Chang SH, Park SH, Cho ML, Choi Y. Why Should We Consider Potential Roles of Oral Bacteria in the Pathogenesis of Sjögren Syndrome? Immune Netw 2022; 22:e32. [PMID: 36081525 PMCID: PMC9433196 DOI: 10.4110/in.2022.22.e32] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 06/16/2022] [Accepted: 06/22/2022] [Indexed: 12/01/2022] Open
Abstract
Sjögren syndrome (SS) is a chronic autoimmune disorder that primarily targets the salivary and lacrimal glands. The pathology of these exocrine glands is characterized by periductal focal lymphocytic infiltrates, and both T cell-mediated tissue injury and autoantibodies that interfere with the secretion process underlie glandular hypofunction. In addition to these adaptive mechanisms, multiple innate immune pathways are dysregulated, particularly in the salivary gland epithelium. Our understanding of the pathogenetic mechanisms of SS has substantially improved during the past decade. In contrast to viral infection, bacterial infection has never been considered in the pathogenesis of SS. In this review, oral dysbiosis associated with SS and evidence for bacterial infection of the salivary glands in SS were reviewed. In addition, the potential contributions of bacterial infection to innate activation of ductal epithelial cells, plasmacytoid dendritic cells, and B cells and to the breach of tolerance via bystander activation of autoreactive T cells and molecular mimicry were discussed. The added roles of bacteria may extend our understanding of the pathogenetic mechanisms and therapeutic approaches for this autoimmune exocrinopathy.
Collapse
Affiliation(s)
- Sung-Ho Chang
- Department of Immunology and Molecular Microbiology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Korea
| | - Sung-Hwan Park
- Divison of Rheumatology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Mi-La Cho
- Department of Medical Life Science, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Youngnim Choi
- Department of Immunology and Molecular Microbiology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Korea
| |
Collapse
|
25
|
Li M, Qi Y, Wang G, Bu S, Chen M, Yu J, Luo T, Meng L, Dai A, Zhou Y, Liu S, Huo X. Proteomic profiling of saliva reveals association of complement system with primary Sjögren's syndrome. IMMUNITY INFLAMMATION AND DISEASE 2021; 9:1724-1739. [PMID: 34516718 PMCID: PMC8589410 DOI: 10.1002/iid3.529] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 08/31/2021] [Accepted: 09/01/2021] [Indexed: 02/06/2023]
Abstract
INTRODUCTION To compare the saliva proteomes of experimental Sjögren's syndrome (ESS) model mice and healthy controls to identify potential diagnostic biomarkers for primary Sjögren's syndrome (pSS). METHODS Proteins were extracted from the saliva of three ESS and three normal control mice using the data-independent acquisition technique. R language was used to identify the differentially expressed proteins (DEPs). Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway analyses were performed to functionally annotate the DEPs. The protein-protein interaction (PPI) network was constructed and the core proteins were identified with the STRING website and Cytoscape software. The concentrations of Serpin family G member 1 (SERPING1), C3, complement factor H (CFH), fibrinogen alpha (FGA), and fibrinogen gamma (FGG) in saliva were determined by ELISA. RESULTS A total of 1722 DEPs were identified in the saliva of the ESS mice relative to the controls, of which 50 showed significantly different expression levels between the two groups. SERPING1, C3, CFH, FGA, and FGG were significantly downregulated, and keratin 4 (Krt4) and transglutaminase 3 (TGM3) were upregulated in the saliva of ESS mice. The PPI network showed that SERPING1, C3, FGG, FGA, TGM3, and hemopexin (HPX) were the core proteins. ELISA results showed that the expression of C3, CFH, FGA, and SERPING1 were significantly downregulated in the saliva of ESS mice. However, the expression of FGG was a little downregulated but with no significant difference. SERPING1, FGG, and FGA may downregulate the complement C3 by inhibiting immune complement system, thereby promoting pSS progression. CONCLUSIONS The salivary proteome of ESS mice was markedly different from that of healthy controls, suggesting that salivary proteomics is a promising noninvasive diagnostic tool for pSS. SERPING1, C3, CFH, FGA, and FGG are potential biomarkers of pSS.
Collapse
Affiliation(s)
- Mingde Li
- Experimental Center of Clinical Research, Scientific Research Department, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China.,Department of Anesthesiology, Second Clinical Medical College, Anhui Medical University, Hefei, Anhui, China
| | - Yajun Qi
- Department of Traditional Chinese Medicine, College of Acupuncture and Massage, Anhui University of traditional Chinese Medicine, Hefei, Anhui, China
| | - Guizhen Wang
- Department of Rheumatology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| | - Su Bu
- Experimental Center of Clinical Research, Scientific Research Department, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Ming Chen
- Department of Anesthesiology, Second Clinical Medical College, Anhui Medical University, Hefei, Anhui, China
| | - Jiahui Yu
- Department of Traditional Chinese Pharmacology, College of Pharmacy, Anhui Medical University, Hefei, Anhui, China
| | - Tianyang Luo
- Department of Anesthesiology, Second Clinical Medical College, Anhui Medical University, Hefei, Anhui, China
| | - Lulu Meng
- Experimental Center of Clinical Research, Scientific Research Department, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Anran Dai
- Experimental Center of Clinical Research, Scientific Research Department, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Yong Zhou
- Experimental Center of Clinical Research, Scientific Research Department, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Shuai Liu
- Experimental Center of Clinical Research, Scientific Research Department, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Xingxing Huo
- Experimental Center of Clinical Research, Scientific Research Department, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
| |
Collapse
|
26
|
Doaré E, Héry-Arnaud G, Devauchelle-Pensec V, Alegria GC. Healthy Patients Are Not the Best Controls for Microbiome-Based Clinical Studies: Example of Sjögren's Syndrome in a Systematic Review. Front Immunol 2021; 12:699011. [PMID: 34394092 PMCID: PMC8358393 DOI: 10.3389/fimmu.2021.699011] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 07/13/2021] [Indexed: 01/03/2023] Open
Abstract
Introduction It has been hypothesized that gut and oral dysbiosis may contribute to the development of primary Sjögren's syndrome (pSS). The aim of this systematic review was to assemble available data regarding the oral and gut microbiota in pSS and to compare them to data from healthy individuals and patients with dry symptoms without a diagnosis of Sjögren's syndrome or lupus disease to identify dysbiosis and discuss the results. Methodology Using the PRISMA guidelines, we systematically reviewed studies that compared the oral and gut microbiota of Sjögren's patients and controls. The PubMed database and Google Scholar were searched. Results Two-hundred and eighty-nine studies were found, and 18 studies were included: 13 referred to the oral microbiota, 4 referred to the gut microbiota, and 1 referred to both anatomical sites. The most frequent controls were healthy volunteers and patients with sicca symptoms. The most common analysis method used was 16S-targeted metagenomics. The results were mostly heterogeneous, and the results regarding diversity were not always in accordance. Dysbiosis in pSS was not confirmed, and reduced salivary secretion seems to explain more microbial changes than the underlying disease. Conclusion These heterogeneous results might be explained by the lack of a standardized methodology at each step of the process and highlight the need for guidelines. Our review provides evidence that sicca patients seem to be more relevant than healthy subjects as a control group.
Collapse
Affiliation(s)
- Elise Doaré
- Rheumatology Department, Reference Centre of Rare Autoimmune Diseases, Cavale Blanche Hospital and Brest University, INSERM UMR 1227, Brest, France
| | - Geneviève Héry-Arnaud
- UMR1078, Génétique, Génomique Fonctionnelle Et Biotechnologies, INSERM, Université de Brest, EFS, IBSAM, Brest, France.,Centre Brestois d'Analyse du Microbiote, Hôpital La Cavale Blanche, CHRU de Brest, Brest, France
| | - Valérie Devauchelle-Pensec
- Rheumatology Department, Reference Centre of Rare Autoimmune Diseases, Cavale Blanche Hospital and Brest University, INSERM UMR 1227, Brest, France
| | - Guillermo Carvajal Alegria
- Rheumatology Department, Reference Centre of Rare Autoimmune Diseases, Cavale Blanche Hospital and Brest University, INSERM UMR 1227, Brest, France
| |
Collapse
|
27
|
Progress in Oral Microbiome Related to Oral and Systemic Diseases: An Update. Diagnostics (Basel) 2021; 11:diagnostics11071283. [PMID: 34359364 PMCID: PMC8306157 DOI: 10.3390/diagnostics11071283] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/09/2021] [Accepted: 07/14/2021] [Indexed: 02/07/2023] Open
Abstract
The human oral microbiome refers to an ecological community of symbiotic and pathogenic microorganisms found in the oral cavity. The oral cavity is an environment that provides various biological niches, such as the teeth, tongue, and oral mucosa. The oral cavity is the gateway between the external environment and the human body, maintaining oral homeostasis, protecting the mouth, and preventing disease. On the flip side, the oral microbiome also plays an important role in the triggering, development, and progression of oral and systemic diseases. In recent years, disease diagnosis through the analysis of the human oral microbiome has been realized with the recent development of innovative detection technology and is overwhelmingly promising compared to the previous era. It has been found that patients with oral and systemic diseases have variations in their oral microbiome compared to normal subjects. This narrative review provides insight into the pathophysiological role that the oral microbiome plays in influencing oral and systemic diseases and furthers the knowledge related to the oral microbiome produced over the past 30 years. A wide range of updates were provided with the latest knowledge of the oral microbiome to help researchers and clinicians in both academic and clinical aspects. The microbial community information can be utilized in non-invasive diagnosis and can help to develop a new paradigm in precision medicine, which will benefit human health in the era of post-metagenomics.
Collapse
|
28
|
Oliveira SG, Nishiyama RR, Trigo CAC, Mattos-Guaraldi AL, Dávila AMR, Jardim R, Aguiar FHB. Core of the saliva microbiome: an analysis of the MG-RAST data. BMC Oral Health 2021; 21:351. [PMID: 34271900 PMCID: PMC8283749 DOI: 10.1186/s12903-021-01719-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 07/07/2021] [Indexed: 11/10/2022] Open
Abstract
Background Oral microbiota is considered as the second most complex in the human body and its dysbiosis can be responsible for oral diseases. Interactions between the microorganism communities and the host allow establishing the microbiological proles. Identifying the core microbiome is essential to predicting diseases and changes in environmental behavior from microorganisms. Methods Projects containing the term “SALIVA”, deposited between 2014 and 2019 were recovered on the MG-RAST portal. Quality (Failed), taxonomic prediction (Unknown and Predicted), species richness (Rarefaction), and species diversity (Alpha) were analyzed according to sequencing approaches (Amplicon sequencing and Shotgun metagenomics). All data were checked for normality and homoscedasticity. Metagenomic projects were compared using the Mann–Whitney U test and Spearman's correlation. Microbiome cores were inferred by Principal Component Analysis. For all statistical tests, p < 0.05 was used. Results The study was performed with 3 projects, involving 245 Amplicon and 164 Shotgun metagenome datasets. All comparisons of variables, according to the type of sequencing, showed significant differences, except for the Predicted. In Shotgun metagenomics datasets the highest correlation was between Rarefaction and Failed (r = − 0.78) and the lowest between Alpha and Unknown (r = − 0.12). In Amplicon sequencing datasets, the variables Rarefaction and Unknown (r = 0.63) had the highest correlation and the lowest was between Alpha and Predicted (r = − 0.03). Shotgun metagenomics datasets showed a greater number of genera than Amplicon. Propionibacterium, Lactobacillus, and Prevotella were the most representative genera in Amplicon sequencing. In Shotgun metagenomics, the most representative genera were Escherichia, Chitinophaga, and Acinetobacter. Conclusions Core of the salivary microbiome and genera diversity are dependent on the sequencing approaches. Available data suggest that Shotgun metagenomics and Amplicon sequencing have similar sensitivities to detect the taxonomic level investigated, although Shotgun metagenomics allows a deeper analysis of the microorganism diversity. Microbiome studies must consider characteristics and limitations of the sequencing approaches. Were identified 20 genera in the core of saliva microbiome, regardless of the health condition of the host. Some bacteria of the core need further study to better understand their role in the oral cavity. Supplementary Information The online version contains supplementary material available at 10.1186/s12903-021-01719-5.
Collapse
Affiliation(s)
- Simone G Oliveira
- Department of Restorative Dentistry, Piracicaba Dental School, State University of Campinas, Av. Limeira, 901, Piracicaba, Brazil.,Faculty of Dentistry, Rio de Janeiro State University, Boulevard 28 de setembro, 157, Rio de Janeiro, Brazil
| | - Rafaela R Nishiyama
- Department of Restorative Dentistry, Piracicaba Dental School, State University of Campinas, Av. Limeira, 901, Piracicaba, Brazil
| | - Claudio A C Trigo
- Department of Restorative Dentistry, Piracicaba Dental School, State University of Campinas, Av. Limeira, 901, Piracicaba, Brazil
| | - Ana Luiza Mattos-Guaraldi
- Laboratory of Diphtheria and Corynebacteria of Clinical Relevance, Faculty of Medical Sciences, Rio de Janeiro State University, Boulevard 28 de setembro, 77, Rio de Janeiro, Brazil
| | - Alberto M R Dávila
- Computational and Systems Biology Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Av. Brasil, 4365, Rio de Janeiro, Brazil
| | - Rodrigo Jardim
- Computational and Systems Biology Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Av. Brasil, 4365, Rio de Janeiro, Brazil.
| | - Flavio H B Aguiar
- Department of Restorative Dentistry, Piracicaba Dental School, State University of Campinas, Av. Limeira, 901, Piracicaba, Brazil
| |
Collapse
|
29
|
Bellando-Randone S, Russo E, Venerito V, Matucci-Cerinic M, Iannone F, Tangaro S, Amedei A. Exploring the Oral Microbiome in Rheumatic Diseases, State of Art and Future Prospective in Personalized Medicine with an AI Approach. J Pers Med 2021; 11:625. [PMID: 34209167 PMCID: PMC8306274 DOI: 10.3390/jpm11070625] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 06/11/2021] [Accepted: 06/28/2021] [Indexed: 12/25/2022] Open
Abstract
The oral microbiome is receiving growing interest from the scientific community, as the mouth is the gateway for numerous potential etiopathogenetic factors in different diseases. In addition, the progression of niches from the mouth to the gut, defined as "oral-gut microbiome axis", affects several pathologies, as rheumatic diseases. Notably, rheumatic disorders (RDs) are conditions causing chronic, often intermittent pain affecting the joints or connective tissue. In this review, we examine evidence which supports a role for the oral microbiome in the etiology and progression of various RDs, including rheumatoid arthritis (RA), Sjogren's syndrome (SS), and systemic lupus erythematosus (SLE). In addition, we address the most recent studies endorsing the oral microbiome as promising diagnostic biomarkers for RDs. Lastly, we introduce the concepts of artificial intelligence (AI), in particular, machine learning (ML) and their general application for understanding the link between oral microbiota and rheumatic diseases, speculating the application of a possible AI approach-based that can be applied to personalized medicine in the future.
Collapse
Affiliation(s)
- Silvia Bellando-Randone
- Department of Clinical and Experimental Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy; (S.B.-R.); (E.R.); (M.M.-C.)
| | - Edda Russo
- Department of Clinical and Experimental Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy; (S.B.-R.); (E.R.); (M.M.-C.)
| | - Vincenzo Venerito
- Rheumatology Unit, Department of Emergency and Organ Transplantations, University of Bari “Aldo Moro”, 70121 Bari, Italy; (V.V.); (F.I.)
| | - Marco Matucci-Cerinic
- Department of Clinical and Experimental Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy; (S.B.-R.); (E.R.); (M.M.-C.)
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases (UnIRAR), IRCCS San Raffaele Hospital, 20132 Milan, Italy
| | - Florenzo Iannone
- Rheumatology Unit, Department of Emergency and Organ Transplantations, University of Bari “Aldo Moro”, 70121 Bari, Italy; (V.V.); (F.I.)
| | - Sabina Tangaro
- Dipartimento Interateneo di Fisica “M. Merlin”, Istituto Nazionale di Fisica Nucleare, Sezione di Bari, 70121 Bari, Italy;
| | - Amedeo Amedei
- Department of Clinical and Experimental Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy; (S.B.-R.); (E.R.); (M.M.-C.)
| |
Collapse
|
30
|
Melchiorre D, Ceccherini MT, Romano E, Cometi L, El-Aoufy K, Bellando-Randone S, Roccotelli A, Bruni C, Moggi-Pignone A, Carboni D, Guiducci S, Lepri G, Tofani L, Pietramellara G, Matucci-Cerinic M. Oral Lactobacillus Species in Systemic Sclerosis. Microorganisms 2021; 9:1298. [PMID: 34203626 PMCID: PMC8232208 DOI: 10.3390/microorganisms9061298] [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: 04/15/2021] [Revised: 06/07/2021] [Accepted: 06/11/2021] [Indexed: 11/16/2022] Open
Abstract
In systemic sclerosis (SSc), the gastrointestinal tract (GIT) plays a central role in the patient's quality of life. The microbiome populates the GIT, where a relationship between the Lactobacillus and gastrointestinal motility has been suggested. In this study, the analysis of oral Lactobacillus species in SSc patients and healthy subjects using culture-independent molecular techniques, together with a review of the literature on microbiota and lactobacilli in SSc, has been carried out. Twenty-nine SSc female patients (mean age 62) and twenty-three female healthy subjects (HS, mean age 57.6) were enrolled and underwent tongue and gum swab sampling. Quantitative PCR was conducted in triplicate using Lactobacillus specific primers rpoB1, rpoB1o and rpoB2 for the RNA-polymerase β subunit gene. Our data show significantly (p = 0.0211) lower LactobacillusspprpoB sequences on the tongue of patients with SSc compared to HS. The mean value of the amount of Lactobacillus ssprpoB gene on the gumsofSSc patients was minor compared to HS. A significant difference between tongue and gums (p = 0.0421) was found in HS but not in SSc patients. In conclusion, our results show a lower presence of Lactobacillus in the oral cavity of SSc patients. This strengthens the hypothesis that Lactobacillus may have both a protective and therapeutic role in SSc patients.
Collapse
Affiliation(s)
- Daniela Melchiorre
- Department of Experimental and Clinical Medicine, Department of Geriatric Medicine, Division of Rheumatology, University of Firenze, 50124 Firenze, Italy; (E.R.); (L.C.); (K.E.-A.); (S.B.-R.); (C.B.); (A.M.-P.); (D.C.); (S.G.); (G.L.); (L.T.); (M.M.-C.)
| | - Maria Teresa Ceccherini
- Department of Agriculture, Food, Environment and Forestry (DAGRI)-University ofFirenze, 50144 Firenze, Italy; (M.T.C.); (A.R.); (G.P.)
| | - Eloisa Romano
- Department of Experimental and Clinical Medicine, Department of Geriatric Medicine, Division of Rheumatology, University of Firenze, 50124 Firenze, Italy; (E.R.); (L.C.); (K.E.-A.); (S.B.-R.); (C.B.); (A.M.-P.); (D.C.); (S.G.); (G.L.); (L.T.); (M.M.-C.)
| | - Laura Cometi
- Department of Experimental and Clinical Medicine, Department of Geriatric Medicine, Division of Rheumatology, University of Firenze, 50124 Firenze, Italy; (E.R.); (L.C.); (K.E.-A.); (S.B.-R.); (C.B.); (A.M.-P.); (D.C.); (S.G.); (G.L.); (L.T.); (M.M.-C.)
| | - Khadija El-Aoufy
- Department of Experimental and Clinical Medicine, Department of Geriatric Medicine, Division of Rheumatology, University of Firenze, 50124 Firenze, Italy; (E.R.); (L.C.); (K.E.-A.); (S.B.-R.); (C.B.); (A.M.-P.); (D.C.); (S.G.); (G.L.); (L.T.); (M.M.-C.)
| | - Silvia Bellando-Randone
- Department of Experimental and Clinical Medicine, Department of Geriatric Medicine, Division of Rheumatology, University of Firenze, 50124 Firenze, Italy; (E.R.); (L.C.); (K.E.-A.); (S.B.-R.); (C.B.); (A.M.-P.); (D.C.); (S.G.); (G.L.); (L.T.); (M.M.-C.)
| | - Angela Roccotelli
- Department of Agriculture, Food, Environment and Forestry (DAGRI)-University ofFirenze, 50144 Firenze, Italy; (M.T.C.); (A.R.); (G.P.)
| | - Cosimo Bruni
- Department of Experimental and Clinical Medicine, Department of Geriatric Medicine, Division of Rheumatology, University of Firenze, 50124 Firenze, Italy; (E.R.); (L.C.); (K.E.-A.); (S.B.-R.); (C.B.); (A.M.-P.); (D.C.); (S.G.); (G.L.); (L.T.); (M.M.-C.)
| | - Alberto Moggi-Pignone
- Department of Experimental and Clinical Medicine, Department of Geriatric Medicine, Division of Rheumatology, University of Firenze, 50124 Firenze, Italy; (E.R.); (L.C.); (K.E.-A.); (S.B.-R.); (C.B.); (A.M.-P.); (D.C.); (S.G.); (G.L.); (L.T.); (M.M.-C.)
| | - Davide Carboni
- Department of Experimental and Clinical Medicine, Department of Geriatric Medicine, Division of Rheumatology, University of Firenze, 50124 Firenze, Italy; (E.R.); (L.C.); (K.E.-A.); (S.B.-R.); (C.B.); (A.M.-P.); (D.C.); (S.G.); (G.L.); (L.T.); (M.M.-C.)
| | - Serena Guiducci
- Department of Experimental and Clinical Medicine, Department of Geriatric Medicine, Division of Rheumatology, University of Firenze, 50124 Firenze, Italy; (E.R.); (L.C.); (K.E.-A.); (S.B.-R.); (C.B.); (A.M.-P.); (D.C.); (S.G.); (G.L.); (L.T.); (M.M.-C.)
| | - Gemma Lepri
- Department of Experimental and Clinical Medicine, Department of Geriatric Medicine, Division of Rheumatology, University of Firenze, 50124 Firenze, Italy; (E.R.); (L.C.); (K.E.-A.); (S.B.-R.); (C.B.); (A.M.-P.); (D.C.); (S.G.); (G.L.); (L.T.); (M.M.-C.)
| | - Lorenzo Tofani
- Department of Experimental and Clinical Medicine, Department of Geriatric Medicine, Division of Rheumatology, University of Firenze, 50124 Firenze, Italy; (E.R.); (L.C.); (K.E.-A.); (S.B.-R.); (C.B.); (A.M.-P.); (D.C.); (S.G.); (G.L.); (L.T.); (M.M.-C.)
| | - Giacomo Pietramellara
- Department of Agriculture, Food, Environment and Forestry (DAGRI)-University ofFirenze, 50144 Firenze, Italy; (M.T.C.); (A.R.); (G.P.)
| | - Marco Matucci-Cerinic
- Department of Experimental and Clinical Medicine, Department of Geriatric Medicine, Division of Rheumatology, University of Firenze, 50124 Firenze, Italy; (E.R.); (L.C.); (K.E.-A.); (S.B.-R.); (C.B.); (A.M.-P.); (D.C.); (S.G.); (G.L.); (L.T.); (M.M.-C.)
| |
Collapse
|
31
|
Verstappen GM, Pringle S, Bootsma H, Kroese FGM. Epithelial-immune cell interplay in primary Sjögren syndrome salivary gland pathogenesis. Nat Rev Rheumatol 2021; 17:333-348. [PMID: 33911236 PMCID: PMC8081003 DOI: 10.1038/s41584-021-00605-2] [Citation(s) in RCA: 150] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/15/2021] [Indexed: 02/08/2023]
Abstract
In primary Sjögren syndrome (pSS), the function of the salivary glands is often considerably reduced. Multiple innate immune pathways are likely dysregulated in the salivary gland epithelium in pSS, including the nuclear factor-κB pathway, the inflammasome and interferon signalling. The ductal cells of the salivary gland in pSS are characteristically surrounded by a CD4+ T cell-rich and B cell-rich infiltrate, implying a degree of communication between epithelial cells and immune cells. B cell infiltrates within the ducts can initiate the development of lymphoepithelial lesions, including basal ductal cell hyperplasia. Vice versa, the epithelium provides chronic activation signals to the glandular B cell fraction. This continuous stimulation might ultimately drive the development of mucosa-associated lymphoid tissue lymphoma. This Review discusses changes in the cells of the salivary gland epithelium in pSS (including acinar, ductal and progenitor cells), and the proposed interplay of these cells with environmental stimuli and the immune system. Current therapeutic options are insufficient to address both lymphocytic infiltration and salivary gland dysfunction. Successful rescue of salivary gland function in pSS will probably demand a multimodal therapeutic approach and an appreciation of the complicity of the salivary gland epithelium in the development of pSS.
Collapse
Affiliation(s)
- Gwenny M Verstappen
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Sarah Pringle
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Hendrika Bootsma
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands.
| | - Frans G M Kroese
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| |
Collapse
|
32
|
Khor B, Snow M, Herrman E, Ray N, Mansukhani K, Patel KA, Said-Al-Naief N, Maier T, Machida CA. Interconnections Between the Oral and Gut Microbiomes: Reversal of Microbial Dysbiosis and the Balance Between Systemic Health and Disease. Microorganisms 2021; 9:496. [PMID: 33652903 PMCID: PMC7996936 DOI: 10.3390/microorganisms9030496] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 02/22/2021] [Accepted: 02/23/2021] [Indexed: 12/13/2022] Open
Abstract
The human microbiota represents a complex array of microbial species that influence the balance between the health and pathology of their surrounding environment. These microorganisms impart important biological benefits to their host, such as immune regulation and resistance to pathogen colonization. Dysbiosis of microbial communities in the gut and mouth precede many oral and systemic diseases such as cancer, autoimmune-related conditions, and inflammatory states, and can involve the breakdown of innate barriers, immune dysregulation, pro-inflammatory signaling, and molecular mimicry. Emerging evidence suggests that periodontitis-associated pathogens can translocate to distant sites to elicit severe local and systemic pathologies, which necessitates research into future therapies. Fecal microbiota transplantation, probiotics, prebiotics, and synbiotics represent current modes of treatment to reverse microbial dysbiosis through the introduction of health-related bacterial species and substrates. Furthermore, the emerging field of precision medicine has been shown to be an effective method in modulating host immune response through targeting molecular biomarkers and inflammatory mediators. Although connections between the human microbiome, immune system, and systemic disease are becoming more apparent, the complex interplay and future innovations in treatment modalities will become elucidated through continued research and cross-disciplinary collaboration.
Collapse
Affiliation(s)
- Brandon Khor
- Academic DMD Program, Oregon Health & Science University, 2730 SW Moody Avenue, Portland, OR 97201, USA; (B.K.); (M.S.); (E.H.); (N.R.); (K.M.); (K.A.P.)
| | - Michael Snow
- Academic DMD Program, Oregon Health & Science University, 2730 SW Moody Avenue, Portland, OR 97201, USA; (B.K.); (M.S.); (E.H.); (N.R.); (K.M.); (K.A.P.)
| | - Elisa Herrman
- Academic DMD Program, Oregon Health & Science University, 2730 SW Moody Avenue, Portland, OR 97201, USA; (B.K.); (M.S.); (E.H.); (N.R.); (K.M.); (K.A.P.)
| | - Nicholas Ray
- Academic DMD Program, Oregon Health & Science University, 2730 SW Moody Avenue, Portland, OR 97201, USA; (B.K.); (M.S.); (E.H.); (N.R.); (K.M.); (K.A.P.)
| | - Kunal Mansukhani
- Academic DMD Program, Oregon Health & Science University, 2730 SW Moody Avenue, Portland, OR 97201, USA; (B.K.); (M.S.); (E.H.); (N.R.); (K.M.); (K.A.P.)
| | - Karan A. Patel
- Academic DMD Program, Oregon Health & Science University, 2730 SW Moody Avenue, Portland, OR 97201, USA; (B.K.); (M.S.); (E.H.); (N.R.); (K.M.); (K.A.P.)
| | - Nasser Said-Al-Naief
- Department of Integrative Biomedical and Diagnostic Sciences, School of Dentistry, Oregon Health & Science University 2730 SW Moody Avenue, Portland, OR 97201, USA; (N.S.-A.-N.); (T.M.)
| | - Tom Maier
- Department of Integrative Biomedical and Diagnostic Sciences, School of Dentistry, Oregon Health & Science University 2730 SW Moody Avenue, Portland, OR 97201, USA; (N.S.-A.-N.); (T.M.)
| | - Curtis A. Machida
- Department of Integrative Biomedical and Diagnostic Sciences, School of Dentistry, Oregon Health & Science University 2730 SW Moody Avenue, Portland, OR 97201, USA; (N.S.-A.-N.); (T.M.)
| |
Collapse
|
33
|
Wang X, Wang J, Guo W, Zhou Y, Sun C, Li Z, Chen L, Pan X. [Characteristics of intestinal flora in patients with primary Sjögren syndrome]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2020; 40:949-957. [PMID: 32895147 DOI: 10.12122/j.issn.1673-4254.2020.07.06] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To investigate changes in intestinal flora in patients with primary Sj?gren syndrome (pSS) and explore the relationship between pSS disease activity and intestinal flora structure. METHODS Fecal samples were collected from 18 female pSS patients, including 9 patients with active disease (group A) and 9 with disease inactivity or low activity (group B), with 10 healthy subjects as the control group. The total bacterial DNA was extracted from the fecal samples for PCR amplification, and Illumina Hiseq 2500 high-throughput sequencing was performed for the v3-v4 region of 16Sr DNA gene to obtain the biological information of the intestinal flora. The intergroup OTU analysis, structural diversity analysis, significant difference analysis and LEFSE analysis were performed with information mining of the literature think tanks. RESULTS The dilution curves generated based on the OTUshannon index for analysis of sample complexity showed that the measured data were relatively complete and could reflect the diversity of the microorganisms in the subjects. Analysis of the Alpha diversity index showed that the Shannon index differed significantly between group A and group B, and the Simpson index differed significantly between group A and group B and between group A and the control group (P < 0.05). Sequence analysis the 3 groups all consisted mainly of 4 phylum (Firmicutes, Bacteroidetes, Actinobacteria, showed that the intestinal flora in and proteobacteria) and 4 genera (finegoldia, Prevotella, Streptococcus, and Corynebacterium_1), all showing no significant differences among the 3 groups (P > 0.05) with the exception of Streptococcus genus, which differed significantly among the 3 groups (P < 0.05). The 16S v3-v4 region in the genus Alloscardovia, Bacteroides, Barnesiella, Butyricicoccus, Facklamia, Faecalibacterium, Lachnospiraceae_FCS020_group, Lachnospiraceae_ND3007_group, Lachnospiraceae_UCG-001, Lachnospirace, Lachnospirace, Ruminococcaceae_UCG-002, Streptococcus and Coprococcus_1 differed significantly among the 3 groups (P < 0.05). The high-dimensional biometrics and genomic characteristics of the intestinal microorganisms differed significantly among the 3 groups (P < 0.05). According to the size of LDA SCORE (effect size), the core flora in group A included the genera Barnesiellaccae, Aerococcaceae, Family-XIII, Bacteroidaceae, Lachnospiraceae_UCG-001, Barnesiella, Facklamia, Alloscardovia, Faecalibacterium and Bacteroides, as compared with the genera Streptococcaceae, Streptococcus, Coprococcus_1, Ruminococcaceae_ucg-002, Lachnospiraceae_FCS020_group, Lachnospiraceae_ucg-004, Lachnospiraceae_ND3007_group, Lachnospiraceae_ucg-008 and Butyricicoccus in the control group. CONCLUSIONS Patients with pSS have significant changes in the diversity of intestinal flora, especially in some specific bacteria in Streptococcu genus and in 16S v3-v4 region of the bacteria. The differences in the core bacteria in the intestinal flora of pSS patients suggest the role of flora structure changes in the pathogenesis of pSS.
Collapse
Affiliation(s)
- Xin Wang
- Department of Rheumatology and Immunology, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China.,Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu 233004, China
| | - Jian Wang
- Department of Rheumatology and Immunology, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China.,Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu 233004, China
| | - Wenjing Guo
- Department of Rheumatology and Immunology, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China.,Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu 233004, China
| | - Ying Zhou
- Department of Rheumatology and Immunology, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China.,Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu 233004, China
| | - Chao Sun
- Department of Rheumatology and Immunology, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China.,Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu 233004, China
| | - Zhijun Li
- Department of Rheumatology and Immunology, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China.,Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu 233004, China
| | - Linjie Chen
- Department of Rheumatology and Immunology, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China.,Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu 233004, China
| | - Xinlan Pan
- Department of Rheumatology and Immunology, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China.,Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu 233004, China
| |
Collapse
|
34
|
Alam J, Lee A, Lee J, Kwon DI, Park HK, Park JH, Jeon S, Baek K, Lee J, Park SH, Choi Y. Dysbiotic oral microbiota and infected salivary glands in Sjögren's syndrome. PLoS One 2020; 15:e0230667. [PMID: 32208441 PMCID: PMC7092996 DOI: 10.1371/journal.pone.0230667] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 03/05/2020] [Indexed: 12/19/2022] Open
Abstract
Key events in the pathogenesis of Sjӧgren syndrome (SS) include the change of salivary gland epithelial cells into antigen-presenting cell-like phenotypes and focal lymphocytic sialadenitis (FLS). However, what triggers these features in SS is unknown. Dysbiosis of the gut and oral microbiomes is a potential environmental factor in SS, but its connection to the etiopathogenesis of SS remains unclear. This study aimed to characterize the oral microbiota in SS and to investigate its potential role in the pathogenesis of SS. Oral bacterial communities were collected by whole mouthwash from control subjects (14 without oral dryness and 11 with dryness) and primary SS patients (8 without oral dryness and 17 with dryness) and were analyzed by pyrosequencing. The SS oral microbiota was characterized by an increased bacterial load and Shannon diversity. Through comparisons of control and SS in combined samples and then separately in non-dry and dry conditions, SS-associated taxa independent of dryness were identified. Three SS-associated species and 2 control species were selected and used to challenge human submandibular gland tumor (HSG) cells. Among the selected SS-associated bacterial species, Prevotella melaninogenica uniquely upregulated the expression of MHC molecules, CD80, and IFNλ in HSG cells. Concomitantly, P. melaninogenica efficiently invaded HSG cells. Sections of labial salivary gland (LSG) biopsies from 8 non-SS subjects and 15 SS patients were subjected to in situ hybridization using universal and P. melaninogenica-specific probes. Ductal cells and the areas of infiltration were heavily infected with bacteria in the LSGs with FLS. Collectively, dysbiotic oral microbiota may initiate the deregulation of SGECs and the IFN signature through bacterial invasion into ductal cells. These findings may provide new insights into the etiopathogenesis of SS.
Collapse
Affiliation(s)
- Jehan Alam
- Departments of Immunology and Molecular Microbiology, Seoul National University School of Dentistry, Seoul, Korea
| | - Ahreum Lee
- Departments of Immunology and Molecular Microbiology, Seoul National University School of Dentistry, Seoul, Korea
| | - Junho Lee
- Departments of Immunology and Molecular Microbiology, Seoul National University School of Dentistry, Seoul, Korea
| | - Dong Il Kwon
- Departments of Immunology and Molecular Microbiology, Seoul National University School of Dentistry, Seoul, Korea
| | - Hee Kyung Park
- Departments of Oral Medicine and Oral Diagnosis, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Korea
| | - Jung-Hyun Park
- Experimental Immunology Branch, National Cancer Institute, NIH, Bethesda, Maryland, United States of America
| | - Sumin Jeon
- Departments of Immunology and Molecular Microbiology, Seoul National University School of Dentistry, Seoul, Korea
| | - Keumjin Baek
- Departments of Immunology and Molecular Microbiology, Seoul National University School of Dentistry, Seoul, Korea
| | - Jennifer Lee
- Division of Rheumatology, Internal medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul, Korea
| | - Sung-Hwan Park
- Division of Rheumatology, Internal medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul, Korea
| | - Youngnim Choi
- Departments of Immunology and Molecular Microbiology, Seoul National University School of Dentistry, Seoul, Korea
| |
Collapse
|
35
|
Martina E, Campanati A, Diotallevi F, Offidani A. Saliva and Oral Diseases. J Clin Med 2020; 9:E466. [PMID: 32046271 PMCID: PMC7074457 DOI: 10.3390/jcm9020466] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 02/03/2020] [Accepted: 02/03/2020] [Indexed: 02/06/2023] Open
Abstract
Saliva is a fascinating biological fluid which has all the features of a perfect diagnostic tool. In fact, its collection is rapid, simple, and noninvasive. Thanks to several transport mechanisms and its intimate contact with crevicular fluid, saliva contains hundreds of proteins deriving from plasma. Advances in analytical techniques have opened a new era-called "salivaomics"-that investigates the salivary proteome, transcriptome, microRNAs, metabolome, and microbiome. In recent years, researchers have tried to find salivary biomarkers for oral and systemic diseases with various protocols and technologies. The review aspires to provide an overall perspective of salivary biomarkers concerning oral diseases such as lichen planus, oral cancer, blistering diseases, and psoriasis. Saliva has proved to be a promising substrate for the early detection of oral diseases and the evaluation of therapeutic response. However, the wide variation in sampling, processing, and measuring of salivary elements still represents a limit for the application in clinical practice.
Collapse
|