1
|
Anton ML, Cardoneanu A, Burlui AM, Mihai IR, Richter P, Bratoiu I, Macovei LA, Rezus E. The Lung in Rheumatoid Arthritis-Friend or Enemy? Int J Mol Sci 2024; 25:6460. [PMID: 38928165 PMCID: PMC11203675 DOI: 10.3390/ijms25126460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 06/09/2024] [Accepted: 06/09/2024] [Indexed: 06/28/2024] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune condition frequently found in rheumatological patients that sometimes raises diagnosis and management problems. The pathogenesis of the disease is complex and involves the activation of many cells and intracellular signaling pathways, ultimately leading to the activation of the innate and acquired immune system and producing extensive tissue damage. Along with joint involvement, RA can have numerous extra-articular manifestations (EAMs), among which lung damage, especially interstitial lung disease (ILD), negatively influences the evolution and survival of these patients. Although there are more and more RA-ILD cases, the pathogenesis is incompletely understood. In terms of genetic predisposition, external environmental factors act and subsequently determine the activation of immune system cells such as macrophages, neutrophils, B and T lymphocytes, fibroblasts, and dendritic cells. These, in turn, show the ability to secrete molecules with a proinflammatory role (cytokines, chemokines, growth factors) that will produce important visceral injuries, including pulmonary changes. Currently, there is new evidence that supports the initiation of the systemic immune response at the level of pulmonary mucosa where the citrullination process occurs, whereby the autoantibodies subsequently migrate from the lung to the synovial membrane. The aim of this paper is to provide current data regarding the pathogenesis of RA-associated ILD, starting from environmental triggers and reaching the cellular, humoral, and molecular changes involved in the onset of the disease.
Collapse
Affiliation(s)
- Maria-Luciana Anton
- Discipline of Rheumatology, Medical Department II, University of Medicine and Pharmacy “Grigore T Popa”, 700115 Iasi, Romania; (M.-L.A.); (A.M.B.); (I.R.M.); (P.R.); (I.B.); (L.A.M.); (E.R.)
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Anca Cardoneanu
- Discipline of Rheumatology, Medical Department II, University of Medicine and Pharmacy “Grigore T Popa”, 700115 Iasi, Romania; (M.-L.A.); (A.M.B.); (I.R.M.); (P.R.); (I.B.); (L.A.M.); (E.R.)
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Alexandra Maria Burlui
- Discipline of Rheumatology, Medical Department II, University of Medicine and Pharmacy “Grigore T Popa”, 700115 Iasi, Romania; (M.-L.A.); (A.M.B.); (I.R.M.); (P.R.); (I.B.); (L.A.M.); (E.R.)
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Ioana Ruxandra Mihai
- Discipline of Rheumatology, Medical Department II, University of Medicine and Pharmacy “Grigore T Popa”, 700115 Iasi, Romania; (M.-L.A.); (A.M.B.); (I.R.M.); (P.R.); (I.B.); (L.A.M.); (E.R.)
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Patricia Richter
- Discipline of Rheumatology, Medical Department II, University of Medicine and Pharmacy “Grigore T Popa”, 700115 Iasi, Romania; (M.-L.A.); (A.M.B.); (I.R.M.); (P.R.); (I.B.); (L.A.M.); (E.R.)
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Ioana Bratoiu
- Discipline of Rheumatology, Medical Department II, University of Medicine and Pharmacy “Grigore T Popa”, 700115 Iasi, Romania; (M.-L.A.); (A.M.B.); (I.R.M.); (P.R.); (I.B.); (L.A.M.); (E.R.)
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Luana Andreea Macovei
- Discipline of Rheumatology, Medical Department II, University of Medicine and Pharmacy “Grigore T Popa”, 700115 Iasi, Romania; (M.-L.A.); (A.M.B.); (I.R.M.); (P.R.); (I.B.); (L.A.M.); (E.R.)
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Elena Rezus
- Discipline of Rheumatology, Medical Department II, University of Medicine and Pharmacy “Grigore T Popa”, 700115 Iasi, Romania; (M.-L.A.); (A.M.B.); (I.R.M.); (P.R.); (I.B.); (L.A.M.); (E.R.)
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| |
Collapse
|
2
|
Filleron A, Cezar R, Fila M, Protsenko N, Van Den Hende K, Jeziorski E, Occean B, Chevallier T, Corbeau P, Tran TA. Regulatory T and B cells in pediatric Henoch-Schönlein purpura: friends or foes? Arthritis Res Ther 2024; 26:52. [PMID: 38365843 PMCID: PMC10870453 DOI: 10.1186/s13075-024-03278-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 01/25/2024] [Indexed: 02/18/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Henoch-Schönlein purpura (HSP) is the most common immunoglobulin A-mediated systemic vasculitis in childhood. We studied immune dysregulation in HSP by analyzing regulatory T (Treg), T helper 3 (Th3), and regulatory B cell (Breg) subpopulations that might intervene in immune activation, IgA production, and HSP clinical manifestations. METHODS This prospective study included 3 groups of children: 30 HSP on acute phase, 30 HSP on remission, and 40 healthy controls (HCs) matched on age. Treg, Breg, and Th3 were analyzed by flow cytometry. Serum immunoglobulin and cytokine levels were quantified by ELISA and Luminex. RESULTS Treg frequencies were higher in acute HSP than in remitting HSP and HCs (6.53% [4.24; 9.21] vs. 4.33% [3.6; 5.66], p = 0.002, and vs. 4.45% [3.01; 6.6], p = 0.003, respectively). Activated Th3 cells (FoxP3 + Th3 cells) tend to be more abundant in HSP than in HCs (78.43% [50.62; 80.84] vs. 43.30% [40.20; 49.32], p = 0.135). Serum IgA, IL-17, and latency-associated peptide (a marker of the anti-inflammatory cytokine TGF-beta production) were significantly and inflammatory cytokines TNF-alpha, IL-1-beta, and IL-6 were non-significantly higher in HSP than HCs. Bregs were identical between the groups, but, in patients with renal impairment, Breg percentage was lower compared to those without. Treg removal in PBMC culture resulted in an increase in IgA production in HSP proving a negative regulatory role of Tregs on IgA production. CONCLUSIONS In pediatric HSP, immune activation persists in spite of an increase in Th3 and Tregs. Th3 could be involved in IgA hyperproduction, inefficiently downregulated by Tregs. Lack of Bregs appears linked to renal impairment.
Collapse
Affiliation(s)
- Anne Filleron
- IRMB, Montpellier University, INSERM U1183, Montpellier, France
- Department of Pediatrics, Nîmes University Hospital, Montpellier University, Service de Pédiatrie, Place du Pr R. Debré, 30029, Nîmes Cedex 9, France
| | - Renaud Cezar
- IRMB, Montpellier University, INSERM U1183, Montpellier, France
- Department of Immunology, Nîmes University Hospital, Montpellier University, Nîmes, France
| | - Marc Fila
- Department of Pediatric Nephrology, Montpellier University Hospital, Montpellier University, Montpellier, France
| | - Nastassja Protsenko
- Department of Pediatrics, Nîmes University Hospital, Montpellier University, Service de Pédiatrie, Place du Pr R. Debré, 30029, Nîmes Cedex 9, France
| | - Kathleen Van Den Hende
- Department of Pediatrics, Nîmes University Hospital, Montpellier University, Service de Pédiatrie, Place du Pr R. Debré, 30029, Nîmes Cedex 9, France
| | - Eric Jeziorski
- Department of Pediatric Infectious Diseases, Montpellier University Hospital, Univ Montpellier, INSERM, EFS, Univ Antilles, Montpellier, France
| | - Bob Occean
- Department of Epidemiology, Medical Statistics and Public Health, Nîmes University Hospital, Montpellier University, Nîmes, France
| | - Thierry Chevallier
- Department of Epidemiology, Medical Statistics and Public Health, Nîmes University Hospital, Montpellier University, Nîmes, France
- UMR 1302 Desbrest Institute of Epidemiology and Public Health, INSERM, University of Montpellier, Montpellier, France
| | - Pierre Corbeau
- Department of Immunology, Nîmes University Hospital, Montpellier University, Nîmes, France
- Institute of Human Genetics, CNRS UMR9002, Montpellier University, Montpellier, France
| | - Tu Anh Tran
- IRMB, Montpellier University, INSERM U1183, Montpellier, France.
- Department of Pediatrics, Nîmes University Hospital, Montpellier University, Service de Pédiatrie, Place du Pr R. Debré, 30029, Nîmes Cedex 9, France.
| |
Collapse
|
3
|
Blenkinsopp HC, Seidler K, Barrow M. Microbial Imbalance and Intestinal Permeability in the Pathogenesis of Rheumatoid Arthritis: A Mechanism Review with a Focus on Bacterial Translocation, Citrullination, and Probiotic Intervention. JOURNAL OF THE AMERICAN NUTRITION ASSOCIATION 2024; 43:59-76. [PMID: 37294082 DOI: 10.1080/27697061.2023.2211129] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 05/01/2023] [Indexed: 06/10/2023]
Abstract
This review aims to investigate the role of intestinal permeability (IP) in rheumatoid arthritis (RA), following the hypotheses that leakage of intestinal microbes can influence increased citrullination of peptides leading to anti-citrullinated protein antibody (ACPA) production and inflammation in RA; and that leaked microbes can migrate to the peripheral joints, leading to immune responses and synovitis in peripheral joints. This review explored the evidence for the link between microbial dysbiosis and increased IP in the inflammatory state in RA, as well as the role of increased citrullination and bacterial translocation in the link between microbiota and immune responses in RA. Furthermore, this research aims to evaluate the potential effect of probiotics on RA symptoms and pathogenesis via proposed mechanisms, including the support of microbial balance and suppression of inflammatory factors in RA. A systematic literature search was conducted in three tranches (review, mechanism, intervention). 71 peer-reviewed papers met the inclusions criteria and are summarized in a narrative analysis. Primary studies were critically appraised, synthesized and their relevance to clinical practice evaluated. Evidence found in this mechanism review consistently supported intestinal dysbiosis and increased IP in arthritis. An altered intestinal microbiome was demonstrated in RA with specific microbes such as Collinsella and Eggerthella correlating with increased IP, mucosal inflammation, and immune responses. Hypercitrullination and ACPA production correlated with arthritic symptoms and intestinal microbes were shown to influence hypercitrullination. Some in vitro and animal studies demonstrated a link between leakage of microbes and bacterial translocation, but further research is needed to elucidate the link between IP and citrullination. Probiotic intervention studies evidenced reductions in inflammatory markers IL-6 and TNFα, associated with proliferation of synovial tissue and pain perception in RA joint inflammation. Despite some conflict in the literature, probiotics may present a promising nutritional intervention in the suppression of both, disease activity and inflammatory markers.Key teaching pointsThere is evidence for a dysbiotic profile of the RA gut with specific RA-associated microbes.Increased intestinal permeability and leakage of PAD enzyme facilitates citrullination of peptides.Hypercitrullination and ACPA production correlate to arthritic signs.Microbial leakage and translocation plays a role in the pathogenesis of RA.Probiotics (e.g. L. Casei 01) may reduce inflammation and ameliorate RA symptoms.
Collapse
Affiliation(s)
- Holly C Blenkinsopp
- The Centre for Nutritional Education and Lifestyle Management (CNELM), Wokingham, UK
| | - Karin Seidler
- The Centre for Nutritional Education and Lifestyle Management (CNELM), Wokingham, UK
| | - Michelle Barrow
- The Centre for Nutritional Education and Lifestyle Management (CNELM), Wokingham, UK
| |
Collapse
|
4
|
Kragsnaes MS, Kjeldsen J, Horn HC, Munk HL, Pedersen JK, Just SA, Ahlquist P, Davidsen JR, Nilsson AC, Röttger R, Kruhøffer M, Marchesi JR, Kristiansen K, Christensen R, Ellingsen T. Response to: 'Correspondence on 'Safety and efficacy of faecal microbiota transplantation for active peripheral psoriatic arthritis: an exploratory randomised placebo-controlled trial'' by McGonagle et al. Ann Rheum Dis 2023; 82:e165. [PMID: 34158373 DOI: 10.1136/annrheumdis-2021-220910] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 06/11/2021] [Indexed: 12/20/2022]
Affiliation(s)
- Maja Skov Kragsnaes
- Rheumatology Research Unit, Department of Rheumatology, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Jens Kjeldsen
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Medical Gastroenterology, Odense University Hospital, Odense, Denmark
| | - Hans Christian Horn
- Rheumatology Research Unit, Department of Rheumatology, Odense University Hospital, Odense, Denmark
| | - Heidi Lausten Munk
- Rheumatology Research Unit, Department of Rheumatology, Odense University Hospital, Odense, Denmark
| | | | - Søren Andreas Just
- Section of Rheumatology, Department of Medicine, Svendborg Hospital, Svendborg, Denmark
| | | | - Jesper Rømhild Davidsen
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- South Danish Center for Interstitial Lung Diseases, Department of Respiratory Medicine, Odense University Hospital, Odense, Denmark
| | | | - Richard Röttger
- Department of Mathematics and Computer Science (IMADA), University of Southern Denmark, Odense, Denmark
| | | | - Julian R Marchesi
- Division of Digestive Diseases, Imperial College London Faculty of Medicine, London, UK
| | - Karsten Kristiansen
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, Copenhagen, Denmark
- Institute of Metagenomics, BGI-Shenzhen, Shenzhen, China
| | - Robin Christensen
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Section for Biostatistics and Evidence-Based Research, the Parker Institute, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
| | - Torkell Ellingsen
- Rheumatology Research Unit, Department of Rheumatology, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
5
|
Rasouli-Saravani A, Jahankhani K, Moradi S, Gorgani M, Shafaghat Z, Mirsanei Z, Mehmandar A, Mirzaei R. Role of microbiota short-chain fatty acid chains in the pathogenesis of autoimmune diseases. Biomed Pharmacother 2023; 162:114620. [PMID: 37004324 DOI: 10.1016/j.biopha.2023.114620] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/27/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023] Open
Abstract
There is emerging evidence that microbiota and its metabolites play an important role in helath and diseases. In this regard, gut microbiota has been found as a crucial component that influences immune responses as well as immune-related disorders such as autoimmune diseases. Gut bacterial dysbiosis has been shown to cause disease and altered microbiota metabolite synthesis, leading to immunological and metabolic dysregulation. Of note, microbiota in the gut produce short-chain fatty acids (SCFAs) such as acetate, butyrate, and propionate, and remodeling in these microbiota metabolites has been linked to the pathophysiology of a number of autoimmune disorders such as type 1 diabetes, multiple sclerosis, inflammatory bowel disease, rheumatoid arthritis, celiac disease, and systemic lupus erythematosus. In this review, we will address the most recent findings from the most noteworthy studies investigating the impact of microbiota SCFAs on various autoimmune diseases.
Collapse
Affiliation(s)
- Ashkan Rasouli-Saravani
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kasra Jahankhani
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shadi Moradi
- Department of Immunology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Melika Gorgani
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Shafaghat
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Mirsanei
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amirreza Mehmandar
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Rasoul Mirzaei
- Venom and Biotherapeutics Molecules Lab, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
6
|
Huang R, Zhang M, Lu Y, Xu D, Liu Y, Jin M, Xian S, Wang S, Tong X, Lu J, Zhang W, Qian W, Tang J, Yang Y, Lu B, Chang Z, Liu X, Ji S. Effects of intestinal microbes on rheumatic diseases: A bibliometric analysis. Front Microbiol 2023; 13:1074003. [PMID: 36699603 PMCID: PMC9870327 DOI: 10.3389/fmicb.2022.1074003] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 12/05/2022] [Indexed: 01/12/2023] Open
Abstract
Background Rheumatic diseases (RD) are a group of multi-system inflammatory autoimmune diseases whose causes are still under study. In the past few decades, researchers have found traces of the association between rheumatic diseases and intestinal microbiota, which can partially explain the pathogenesis of rheumatic diseases. We aimed to describe the research trend and main divisions on how gut flora interreacts with rheumatic diseases, and discussed about the possible clinical applications. Methods We analyzed bibliometric data from the Web of Science core collection (dated 15th May 2022). Biblioshiny R language software packages (bibliometrix) were used to obtain the annual publication and citations, core sources according to Bradford's law, and country collaboration map. We designed and verified the keyword co-occurrence network and strategic diagram with the help of VOSviewer and CiteSpace, subdivided the research topic into several themes and identified research dimensions. The tables of most local cited documents and core sources were processed manually. Furthermore, the Altmetric Attention Score and the annual Altmetric Top 100 were applied to analyze the annual publication and citation. Results From a total of 541 documents, we found that the overall trend of annual publication and citation is increasing. The major research method is to compare the intestinal microbial composition of patients with certain rheumatic disease and that of the control group to determine microbial alterations related to the disease's occurrence and development. According to Bradford's law, the core sources are Arthritis and Rheumatology, Annals of the Rheumatic Diseases, Current Opinion in Rheumatology, Nutrients, Rheumatology, and Journal of Rheumatology. Since 1976, 101 countries or regions have participated in studies of rheumatology and intestinal microbes. The United States ranks at the top and has the broadest academic association with other countries. Five themes were identified, including the pivotal role of inflammation caused by intestinal bacteria in the rheumatic pathogenesis, the close relationship between rheumatic diseases and inflammatory bowel disease, immunoregulation mechanism as a mediator of the interaction between rheumatic diseases and gut flora, dysbiosis and decreased diversity in intestine of patients with rheumatic diseases, and the influence of oral flora on rheumatic diseases. Additionally, four research dimensions were identified, including pathology, treatment, disease, and experiments. Conclusion Studies on rheumatic diseases and the intestinal microbiota are growing. Attention should be paid to the mechanism of their interaction, such as the microbe-immune-RD crosstalk. Hopefully, the research achievements can be applied to diseases' prevention, diagnosis, and treatment, and our work can contribute to the readers' future research.
Collapse
Affiliation(s)
- Runzhi Huang
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China,Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, Shanghai, China
| | - Mengyi Zhang
- School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yuwei Lu
- School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Dayuan Xu
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China,Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, Shanghai, China
| | - Yifan Liu
- School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Minghao Jin
- School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shuyuan Xian
- School of Medicine, Tongji University, Shanghai, China
| | - Siqiao Wang
- School of Medicine, Tongji University, Shanghai, China
| | - Xirui Tong
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China,Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, Shanghai, China
| | - Jianyu Lu
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China,Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, Shanghai, China
| | - Wei Zhang
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China,Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, Shanghai, China
| | - Weijin Qian
- School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jieling Tang
- School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yiting Yang
- School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Bingnan Lu
- School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhengyan Chang
- Department of Pathology, School of Medicine, Shanghai Tenth People’s Hospital, Tongji University, Shanghai, China,*Correspondence: Zhengyan Chang,
| | - Xin Liu
- Department of Rheumatology and Immunology, Second Affiliated Hospital of Naval Medical University, Shanghai, China,Xin Liu,
| | - Shizhao Ji
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China,Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, Shanghai, China,Shizhao Ji,
| |
Collapse
|
7
|
Chelpachenko OE, Danilova EI, Perunova NB, Ivanova EV, Nikiforov IA, Chainikova IN, Bekpergenova AV, Bondarenko TA. Quantitative method for the determination of antibiotic-resistant gut bacterial strains for the early diagnosis of chronic arthritis. Klin Lab Diagn 2022; 67:525-529. [PMID: 36099462 DOI: 10.51620/0869-2084-2022-67-9-525-529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Based on the clinical and microbiological monitoring of two groups of children aged 3 to 17 years with acute (n=78) and chronic (n=46) course of reactive arthritis (ReA), a method for early diagnosis of chronic arthritis was developed by determining the number of antibiotic-resistant coprostrains in patients with ReA, characterized by the absence of the need to isolate a pure culture of the pathogen and its identification; inoculation of faeces at a dilution of 10-5 on solid 1.5% GRM-agar with an antibacterial agents used in the treatment of a particular patient, at a minimum inhibitory concentration in the resistance range, followed by incubation and counting of the colonies of microorganisms grown on the plate. A significant relationship between the number of antibiotic-resistant gut bacterial strains and the course of arthritis (acute, chronic) was revealed, and the borderline value of the number of antibiotic-resistant gut bacterial strains was determined - 5×103 CFU/g, which allows differentiating the acute course from the chronic one: in the acute course< 5×103 CFU/g, with chronic - ≥ 5×103 CFU/g. The method allows, at the stage of completion of anti-inflammatory therapy in the active phase of the disease, to identify a risk group for the development of a chronic course of arthritis among patients with ReA, which can contribute to timely therapeutic measures aimed at preventing recurrence of the disease and making the patient disabled.
Collapse
Affiliation(s)
- O E Chelpachenko
- Institute of Cellular and Intracellular Symbiosis, Ural Department of the Russian Academy of Sciences
| | | | - N B Perunova
- Institute of Cellular and Intracellular Symbiosis, Ural Department of the Russian Academy of Sciences
| | - E V Ivanova
- Institute of Cellular and Intracellular Symbiosis, Ural Department of the Russian Academy of Sciences
- Orenburg State Medical University
| | - I A Nikiforov
- Institute of Cellular and Intracellular Symbiosis, Ural Department of the Russian Academy of Sciences
| | - I N Chainikova
- Institute of Cellular and Intracellular Symbiosis, Ural Department of the Russian Academy of Sciences
- Orenburg State Medical University
| | | | - T A Bondarenko
- Institute of Cellular and Intracellular Symbiosis, Ural Department of the Russian Academy of Sciences
| |
Collapse
|
8
|
Li B, Yang B, Liu X, Zhao J, Ross RP, Stanton C, Zhang H, Chen W. Microbiota-assisted therapy for systemic inflammatory arthritis: advances and mechanistic insights. Cell Mol Life Sci 2022; 79:470. [PMID: 35932328 PMCID: PMC11072763 DOI: 10.1007/s00018-022-04498-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/18/2022] [Accepted: 07/19/2022] [Indexed: 12/22/2022]
Abstract
Research on the influence of gut microbiota on systemic inflammatory arthritis has exploded in the past decade. Gut microbiota changes may be a crucial regulatory component in systemic inflammatory arthritis. As a result of advancements in the field, microbiota-assisted therapy has evolved, but this discipline is still in its infancy. Consequently, we review the limitations of current systemic inflammatory arthritis treatment, analyze the connection between the microbiota and arthritis, and summarize the research progress of microbiota regulating systemic inflammatory arthritis and the further development aspects of microbiota-assisted therapy. Finally, the partial mechanisms of microbiota-assisted therapy of systemic inflammatory arthritis are being discussed. In general, this review summarizes the current progress, challenges, and prospects of microbiota-assisted therapy for systemic inflammatory arthritis and points out the direction for the development of microbiota-assisted therapy in the future.
Collapse
Affiliation(s)
- Bowen Li
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi, 214122, China
| | - Bo Yang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China.
- School of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi, 214122, China.
- International Joint Research Center for Probiotics and Gut Health, Jiangnan University, Wuxi, Jiangsu, China.
| | - Xiaoming Liu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi, 214122, China
- International Joint Research Center for Probiotics and Gut Health, Jiangnan University, Wuxi, Jiangsu, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi, 214122, China
- International Joint Research Center for Probiotics and Gut Health, Jiangnan University, Wuxi, Jiangsu, China
| | - R Paul Ross
- International Joint Research Center for Probiotics and Gut Health, Jiangnan University, Wuxi, Jiangsu, China.
- APC Microbiome Ireland, University College Cork, Cork, Ireland.
| | - Catherine Stanton
- International Joint Research Center for Probiotics and Gut Health, Jiangnan University, Wuxi, Jiangsu, China
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi, 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, China
- Wuxi Translational Medicine Research Center and Jiangsu Translational Medicine Research Institute Wuxi Branch, Wuxi, China
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China.
- School of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi, 214122, China.
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, China.
- Beijing Innovation Center of Food Nutrition and Human Health, Beijing Technology and Business University (BTBU), Beijing, China.
| |
Collapse
|
9
|
Cabana-Puig X, Mu Q, Lu R, Swartwout B, Abdelhamid L, Zhu J, Prakash M, Cecere TE, Wang Z, Callaway S, Sun S, Reilly CM, Ahmed S, Luo XM. Lactobacillus spp. act in synergy to attenuate splenomegaly and lymphadenopathy in lupus-prone MRL/ lpr mice. Front Immunol 2022; 13:923754. [PMID: 35967418 PMCID: PMC9368192 DOI: 10.3389/fimmu.2022.923754] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 07/06/2022] [Indexed: 11/27/2022] Open
Abstract
Commensal bacteria and the immune system have a close and strong relationship that maintains a balance to control inflammation. Alterations of the microbiota, known as dysbiosis, can direct reactivity to self-antigens not only in the intestinal mucosa but also at the systemic level. Our laboratory previously reported gut dysbiosis, particularly lower abundance of bacteria in the family Lactobacillaceae, in lupus-prone MRL/lpr mice, a model of systemic autoimmunity. Restoring the microbiota with a mix of 5 different Lactobacillus species (spp.), L. reuteri, L. oris, L. johnsonii, L. gasseri and L. rhamnosus, attenuated lupus-liked clinical signs, including splenomegaly and lymphadenopathy. However, our understanding of the mechanism was limited. In this study, we first investigated the effects of individual species. Surprisingly, none of the species individually recapitulated the benefits of the mix. Instead, Lactobacillus spp. acted synergistically to attenuate splenomegaly and renal lymphadenopathy through secreted factors and a CX3CR1-dependent mechanism. Interestingly, oral administration of MRS broth exerted the same benefits likely through increasing the relative abundance of endogenous Lactobacillus spp. Mechanistically, we found increased percentages of FOXP3-negative type 1 regulatory T cells with administration of the mix in both spleen and mesenteric lymph nodes. In addition, oral gavage of Lactobacillus spp. decreased the percentage of central memory T cells while increasing that of effector memory T cells in the lymphoid organs. Furthermore, a decreased percentage of double negative T cells was observed in the spleen with the mix. These results suggest that Lactobacillus spp. might act on T cells to attenuate splenomegaly and lymphadenopathy. Together, this study advances our understanding of how Lactobacillus spp. attenuate lupus in MRL/lpr mice. The synergistic action of these bacteria suggests that multiple probiotic bacteria in combination may dampen systemic autoimmunity and benefit lupus patients.
Collapse
Affiliation(s)
- Xavier Cabana-Puig
- Department of Biomedical Sciences and Pathobiology, College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| | - Qinghui Mu
- Department of Biomedical Sciences and Pathobiology, College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| | - Ran Lu
- Department of Biomedical Sciences and Pathobiology, College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| | - Brianna Swartwout
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Roanoke, VA, United States
| | - Leila Abdelhamid
- Department of Biomedical Sciences and Pathobiology, College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| | - Jing Zhu
- Department of Biomedical Sciences and Pathobiology, College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| | - Meeta Prakash
- Carilion School of Medicine, Virginia Tech, Roanoke, VA, United States
| | - Thomas E. Cecere
- Department of Biomedical Sciences and Pathobiology, College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| | - Zhuang Wang
- Department of Biomedical Sciences and Pathobiology, College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| | - Sabrina Callaway
- Department of Biomedical Sciences and Pathobiology, College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| | - Sha Sun
- Department of Development and Cell Biology, University of California, Irvine, Irvine, CA, United States
| | | | - S. Ansar Ahmed
- Department of Biomedical Sciences and Pathobiology, College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| | - Xin M. Luo
- Department of Biomedical Sciences and Pathobiology, College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| |
Collapse
|
10
|
Han Y, Sheng Q, Fang Y. Exploring the Spatial Distribution of Rheumatic Diseases and Its Correlation With Temperature and Humidity Among Middle-Aged and Elderly Adults in China. Int J Public Health 2022; 67:1604782. [PMID: 35936998 PMCID: PMC9351402 DOI: 10.3389/ijph.2022.1604782] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 06/24/2022] [Indexed: 11/13/2022] Open
Abstract
Objectives: This study aimed to analyze the prevalence of rheumatic diseases and its correlation with temperature and humidity among middle-aged and elderly adults in China from a spatial perspective.Methods: Data on rheumatic diseases among middle-aged and elderly adults were sourced from the 2018 China Health and Retirement Longitudinal Study (CHARLS). Moran’s I was applied to explore the spatial autocorrelation of rheumatic diseases. Spatial lag model (SLM) was established to probe the correlation between rheumatic diseases and temperature and humidity.Results: The age-standardized prevalence of rheumatic diseases was 33.2% for middle-aged and elderly adults in China, varying from 12.0% to 51.4% depending on regions. The Global Moran’s I was 0.506 (p = 0.001). Average temperature had negative correlation while average relative humidity had positive correlation with age-standardized prevalence of rheumatic diseases in the SLM.Conclusion: The age-standardized prevalence of rheumatic diseases of middle-aged and elderly adults showed spatial autocorrelation in China. We recommend taking measures to prevent rheumatic diseases for the middle-aged and elderly adults, especially for those living in cold and humid regions.
Collapse
|
11
|
MicrobiomeGWAS: A Tool for Identifying Host Genetic Variants Associated with Microbiome Composition. Genes (Basel) 2022; 13:genes13071224. [PMID: 35886007 PMCID: PMC9317577 DOI: 10.3390/genes13071224] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 06/30/2022] [Accepted: 07/01/2022] [Indexed: 11/17/2022] Open
Abstract
The microbiome is the collection of all microbial genes and can be investigated by sequencing highly variable regions of 16S ribosomal RNA (rRNA) genes. Evidence suggests that environmental factors and host genetics may interact to impact human microbiome composition. Identifying host genetic variants associated with human microbiome composition not only provides clues for characterizing microbiome variation but also helps to elucidate biological mechanisms of genetic associations, prioritize genetic variants, and improve genetic risk prediction. Since a microbiota functions as a community, it is best characterized by β diversity; that is, a pairwise distance matrix. We develop a statistical framework and a computationally efficient software package, microbiomeGWAS, for identifying host genetic variants associated with microbiome β diversity with or without interacting with an environmental factor. We show that the score statistics have positive skewness and kurtosis due to the dependent nature of the pairwise data, which makes p-value approximations based on asymptotic distributions unacceptably liberal. By correcting for skewness and kurtosis, we develop accurate p-value approximations, whose accuracy was verified by extensive simulations. We exemplify our methods by analyzing a set of 147 genotyped subjects with 16S rRNA microbiome profiles from non-malignant lung tissues. Correcting for skewness and kurtosis eliminated the dramatic deviation in the quantile–quantile plots. We provided preliminary evidence that six established lung cancer risk SNPs were collectively associated with microbiome composition for both unweighted (p = 0.0032) and weighted (p = 0.011) UniFrac distance matrices. In summary, our methods will facilitate analyzing large-scale genome-wide association studies of the human microbiome.
Collapse
|
12
|
Zorgetto-Pinheiro VA, Machate DJ, Figueiredo PS, Marcelino G, Hiane PA, Pott A, Guimarães RDCA, Bogo D. Omega-3 Fatty Acids and Balanced Gut Microbiota on Chronic Inflammatory Diseases: A Close Look at Ulcerative Colitis and Rheumatoid Arthritis Pathogenesis. J Med Food 2022; 25:341-354. [PMID: 35438557 DOI: 10.1089/jmf.2021.0012] [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: 12/12/2022] Open
Abstract
The aim of this article was to review experimental and clinical studies regarding the use of omega-3 fatty acids on the prevention and control of chronic inflammatory diseases with autoimmune background through the gut microbiota modulation. For this, natural omega-3 sources are presented emphasizing the importance of a healthy diet for the body's homeostasis and the enzymatic processes that these fatty acids go through once inside the body. The pathogenesis of ulcerative colitis and rheumatoid arthritis are revisited under the light of the gut microbiota dysbiosis approach and how those fatty acids are able to prevent and control these two pathological conditions that are responsible for the global chronic burden and functional disability and life-threatening comorbidities if not treated properly. As a matter of reflection, as we are living a pandemic crisis owing to COVID-19 infection, we present the potential of omega-3 in preventing a poor prognosis once they contribute to balancing the immune system modulation the inflammatory process.
Collapse
Affiliation(s)
- Verônica Assalin Zorgetto-Pinheiro
- Graduate Program in Health and Development in the Central-West Region, Federal University of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil
| | - David Johane Machate
- Group of Spectroscopy and Bioinformatics Applied Biodiversity and Health (GEBABS), Graduate Program in Science of Materials, Federal University of Mato Grosso do Sul, Mato Grosso do Sul, Brazil
| | - Priscila Silva Figueiredo
- Graduate Program in Health and Development in the Central-West Region, Federal University of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil
| | - Gabriela Marcelino
- Graduate Program in Health and Development in the Central-West Region, Federal University of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil
| | - Priscila Aiko Hiane
- Graduate Program in Health and Development in the Central-West Region, Federal University of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil
| | - Arnildo Pott
- Graduate Program in Biotechnology and Biodiversity in the Central-West Region of Brazil, Federal University of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil
| | - Rita de Cássia Avellaneda Guimarães
- Graduate Program in Health and Development in the Central-West Region, Federal University of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil
| | - Danielle Bogo
- Graduate Program in Health and Development in the Central-West Region, Federal University of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil
| |
Collapse
|
13
|
Abstract
Cardiovascular diseases (CVDs) still remain the leading concern of global health, accounting for approximately 17.9 million deaths in 2016. The pathogenetic mechanisms of CVDs are multifactorial and incompletely understood. Recent evidence has shown that alterations in the gut microbiome and its associated metabolites may influence the pathogenesis and progression of CVDs such as atherosclerosis, heart failure, hypertension, and arrhythmia, yet the underlying links are not fully elucidated. Owing to the progress in next-generation sequencing techniques and computational strategies, researchers now are available to explore the emerging links to the genomes, transcriptomes, proteomes, and metabolomes in parallel meta-omics approaches, presenting a panoramic vista of culture-independent microbial investigation. This review aims to outline the characteristics of meta-omics pipelines and provide a brief overview of current applications in CVDs studies which can be practical for addressing crucial knowledge gaps in this field, as well as to shed its light on cardiovascular risk biomarkers and therapeutic intervention in the near future.
Collapse
Affiliation(s)
- Jing Xu
- State Key Laboratory of Cardiovascular Diseases, Fuwai Hospital & National Center for Cardiovascular Diseases, Beijing, China,Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yuejin Yang
- State Key Laboratory of Cardiovascular Diseases, Fuwai Hospital & National Center for Cardiovascular Diseases, Beijing, China,Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China,CONTACT Yuejin Yang State Key Laboratory of Cardiovascular Disease, Fuwai Hospital & National Center for Cardiovascular Disease, Beijing, China; Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| |
Collapse
|
14
|
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: 1.3] [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
|
15
|
Jia X, Zhai T, Qu C, Ye J, Zhao J, Liu X, Zhang JA, Qian Q. Metformin Reverses Hashimoto's Thyroiditis by Regulating Key Immune Events. Front Cell Dev Biol 2021; 9:685522. [PMID: 34124070 PMCID: PMC8193849 DOI: 10.3389/fcell.2021.685522] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 05/05/2021] [Indexed: 11/13/2022] Open
Abstract
Background Hashimoto's thyroiditis (HT) is a common autoimmune disease characterized by high levels of thyroid peroxidase antibody (TPOAb) and thyroid globulin antibody (TgAb) as well as infiltration of lymphocytes in thyroid. In recent years, metformin has been proven to be effective in a variety of autoimmune diseases, such as systemic lupus erythematosus, rheumatoid arthritis and multiple sclerosis. Methods This study systematically explored the therapeutic effect of metformin on HT and its underlying mechanism by comprehensively utilizing methods including animal model, in vitro cell culture and differentiation, mRNA sequencing and 16S rRNA sequencing. Findings We found that metformin indeed had a therapeutic effect on mice with HT mainly by reducing TgAb and lymphocyte infiltration in thyroid tissue. In addition, metformin also significantly suppressed the number and function of Th17 cells and M1 macrophages polarization in HT mice. Furthermore, metformin can inhibit the differentiation and function of Th17 in vitro. The results of mRNA sequencing of thyroid tissue illustrated that the therapeutic effect of metformin on HT was mainly achieved by regulating immune pathways. 16S RNA sequencing of the intestinal flora found that the intestinal flora of HT mice differs significantly from that of the normal mice and also were altered by metformin treatment. Interpretation These experiments provided a preliminary theoretical basis for the clinical application of metformin in the treatment of HT.
Collapse
Affiliation(s)
- Xi Jia
- Department of Endocrinology, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Tianyu Zhai
- Department of Endocrinology, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Chunjie Qu
- Shanghai Pudong New Area Center for Disease Control, Shanghai, China
| | - Jianjun Ye
- Shanghai Kangqiao Community Health Service Center, Shanghai, China
| | - Jing Zhao
- Department of Endocrinology, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Xuerong Liu
- Department of Endocrinology, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Jin-An Zhang
- Department of Endocrinology, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Qiaohui Qian
- Department of Endocrinology, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| |
Collapse
|
16
|
Xiao S, Zhang G, Jiang C, Liu X, Wang X, Li Y, Cheng M, Lv H, Xian F, Guo X, Tan Y. Deciphering Gut Microbiota Dysbiosis and Corresponding Genetic and Metabolic Dysregulation in Psoriasis Patients Using Metagenomics Sequencing. Front Cell Infect Microbiol 2021; 11:605825. [PMID: 33869074 PMCID: PMC8047475 DOI: 10.3389/fcimb.2021.605825] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 03/09/2021] [Indexed: 12/21/2022] Open
Abstract
Background Increasing evidence has shown that alterations in the intestinal microbiota play an important role in the pathogenesis of psoriasis. The existing relevant studies focus on 16S rRNA gene sequencing, but in-depth research on gene functions and comprehensive identification of microbiota is lacking. Objectives To comprehensively identify characteristic gut microbial compositions, genetic functions and relative metabolites of patients with psoriasis and to reveal the potential pathogenesis of psoriasis. Methods DNA was extracted from the faecal microbiota of 30 psoriatic patients and 15 healthy subjects, and metagenomics sequencing and bioinformatic analyses were performed. The Kyoto Encyclopedia of Genes and Genomes (KEGG) database, cluster of orthologous groups (COG) annotations, and metabolic analyses were used to indicate relative target genes and pathways to reveal the pathogenesis of psoriasis. Results Compared with healthy individuals, the gut microbiota of psoriasis patients displayed an alteration in microbial taxa distribution, but no significant difference in microbial diversity. A distinct gut microbial composition in patients with psoriasis was observed, with an increased abundance of the phyla Firmicutes, Actinobacteria and Verrucomicrobia and genera Faecalibacterium, Bacteroides, Bifidobacterium, Megamonas and Roseburia and a decreased abundance of the phyla Bacteroidetes, Euryarchaeota and Proteobacteria and genera Prevotella, Alistipes, and Eubacterium. A total of 134 COGs were predicted with functional analysis, and 15 KEGG pathways, including lipopolysaccharide (LPS) biosynthesis, WNT signaling, apoptosis, bacterial secretion system, and phosphotransferase system, were significantly enriched in psoriasis patients. Five metabolites, hydrogen sulfide (H2S), isovalerate, isobutyrate, hyaluronan and hemicellulose, were significantly dysregulated in the psoriatic cohort. The dysbiosis of gut microbiota, enriched pathways and dysregulated metabolites are relevant to immune and inflammatory response, apoptosis, the vascular endothelial growth factor (VEGF) signaling pathway, gut-brain axis and brain-skin axis that play important roles in the pathogenesis of psoriasis. Conclusions A clear dysbiosis was displayed in the gut microbiota profile, genetic functions and relative metabolites of psoriasis patients. This study is beneficial for further understanding the inflammatory pathogenesis of psoriasis and could be used to develop microbiome-based predictions and therapeutic approaches.
Collapse
Affiliation(s)
- Shiju Xiao
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,Graduate School, Capital Medical University, Beijing, China
| | - Guangzhong Zhang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Chunyan Jiang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Xin Liu
- Puyang Hospital of Traditional Chinese Medicine, Puyang, China
| | - Xiaoxu Wang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,Graduate School, Capital Medical University, Beijing, China
| | - Yafan Li
- Beijing University of Chinese Medicine, Beijing, China
| | - Meijiao Cheng
- Beijing QuantiHealth Technology Co., Ltd, Beijing, China
| | - Hongpeng Lv
- Beijing University of Chinese Medicine, Beijing, China
| | - Fuyang Xian
- Beijing University of Chinese Medicine, Beijing, China
| | - Xinwei Guo
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,Graduate School, Capital Medical University, Beijing, China.,Beijing Institute of Traditional Chinese Medicine, Beijing, China
| | - Yong Tan
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
17
|
New Studies of Pathogenesis of Rheumatoid Arthritis with Collagen-Induced and Collagen Antibody-Induced Arthritis Models: New Insight Involving Bacteria Flora. Autoimmune Dis 2021; 2021:7385106. [PMID: 33833871 PMCID: PMC8016593 DOI: 10.1155/2021/7385106] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 12/30/2020] [Accepted: 03/04/2021] [Indexed: 12/23/2022] Open
Abstract
Much public research suggests that autoimmune diseases such as rheumatoid arthritis (RA) are induced by aberrant “self” immune responses attacking autologous tissues and organ components. However, recent studies have reported that autoimmune diseases may be triggered by dysbiotic composition changes of the intestinal bacteria and an imbalance between these bacteria and intestinal immune systems. However, there are a few solid concepts or methods to study the putative involvement and relationship of these inner environmental factors in RA pathogenesis. Fortunately, Collagen-Induced Arthritis (CIA) and Collagen Antibody-Induced Arthritis (CAIA) models have been widely used as animal models for studying the pathogenesis of RA. In addition to RA, these models can be extensively used as animal models for studying complicated hypotheses in many diseases. In this review, we introduce some basic information about the CIA and CAIA models as well as how to apply these models effectively to investigate relationships between the pathogenesis of autoimmune diseases, especially RA, and the dysbiosis of intestinal bacterial flora.
Collapse
|
18
|
The Interrelationships between Intestinal Permeability and Phlegm Syndrome and Therapeutic Potential of Some Medicinal Herbs. Biomolecules 2021; 11:biom11020284. [PMID: 33671865 PMCID: PMC7918952 DOI: 10.3390/biom11020284] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 02/06/2021] [Accepted: 02/10/2021] [Indexed: 02/06/2023] Open
Abstract
The gastrointestinal (GI) tract has an intriguing and critical role beyond digestion in both modern and complementary and alternative medicine (CAM), as demonstrated by its link with the immune system. In this review, we attempted to explore the interrelationships between increased GI permeability and phlegm, an important pathological factor in CAM, syndrome, and therapeutic herbs for two disorders. The leaky gut and phlegm syndromes look considerably similar with respect to related symptoms, diseases, and suitable herbal treatment agents, including phytochemicals even though limitations to compare exist. Phlegm may be spread throughout the body along with other pathogens via the disruption of the GI barrier to cause several diseases sharing some parts of symptoms, diseases, and mechanisms with leaky gut syndrome. Both syndromes are related to inflammation and gut microbiota compositions. Well-designed future research should be conducted to verify the interrelationships for evidence based integrative medicine to contribute to the promotion of public health. In addition, systems biology approaches should be adopted to explore the complex synergistic effects of herbal medicine and phytochemicals on conditions associated with phlegm and leaky gut syndromes.
Collapse
|
19
|
Dysregulation of the gut-brain-skin axis and key overlapping inflammatory and immune mechanisms of psoriasis and depression. Biomed Pharmacother 2021; 137:111065. [PMID: 33540138 DOI: 10.1016/j.biopha.2020.111065] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 11/14/2020] [Accepted: 11/20/2020] [Indexed: 12/21/2022] Open
Abstract
The occurrence, progression and recurrence of psoriasis are thought to be related to mood and psychological disorders such as depression. Psoriasis can lead to depression, and depression, in turn, exacerbates psoriasis. No specific mechanism can explain the association between psoriasis and depression. The gut-brain-skin axis has been used to explain correlations among the gut microbiota, emotional states and systemic and skin inflammation, and this axis may be associated with overlapping mechanisms between psoriasis and depression. Therefore, in the context of the gut-brain-skin axis, we systematically summarized and comparatively analysed the inflammatory and immune mechanisms of psoriasis and depression and illustrated the dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis and the gut microbiota. This review provides a theoretical basis and new targets for the treatment of psoriasis and depression.
Collapse
|
20
|
Xin L, He F, Li S, Zhou ZX, Ma XL. Intestinal microbiota and juvenile idiopathic arthritis: current understanding and future prospective. World J Pediatr 2021; 17:40-51. [PMID: 32533534 DOI: 10.1007/s12519-020-00371-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 05/24/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND Juvenile idiopathic arthritis (JIA) characterized by arthritis of unknown origin is the most common childhood chronic rheumatic disease, caused by both host genetic factors and environmental triggers. Recent evidence has mounted to focus on the intestinal microbiota, a potentially recognized set of environmental triggers affecting JIA development. Here we offer an overview of recently published animal and human studies that support the impact of intestinal microbiota in JIA. DATA SOURCES We searched PubMed for animal and human studies publications with the search terms "intestinal microbiota or gut microbiota" and "juvenile idiopathic arthritis or juvenile chronic arthritis or juvenile rheumatoid arthritis or childhood rheumatoid arthritis or pediatric rheumatoid arthritis". RESULTS Several comparative studies have demonstrated that intestinal microbial alterations might be triggers in disease pathogenesis. Alternatively, a slice of studies has suggested environmental triggers in early life might disrupt intestinal microbial colonization, including cesarean section, formula feeding, and antibiotic exposure. Aberrant intestinal microbiota may influence the development of JIA by mediating host immune programming and by altering mucosal permeability. CONCLUSIONS Specific microbial factors may contribute to the pathogenesis of JIA. Intensive studies, however, are warranted to investigate the causality between intestinal dysbiosis and JIA and the mechanisms behind these epidemiologic relationships. Studies are also needed to design the best interventional administrations to restore balanced intestinal microbial communities.
Collapse
Affiliation(s)
- Le Xin
- Gynecological Minimal Invasive Center, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Feng He
- Department of Biochemistry and Immunology, Capital Institute of Pediatrics, Yabao Road No. 2, Chaoyang District, Beijing, China
| | - Sen Li
- Department of Biochemistry and Immunology, Capital Institute of Pediatrics, Yabao Road No. 2, Chaoyang District, Beijing, China
| | - Zhi-Xuan Zhou
- Department of Rheumatology, Capital Institute of Pediatrics, Beijing, China
| | - Xiao-Lin Ma
- Department of Rheumatology, Capital Institute of Pediatrics, Beijing, China.
| |
Collapse
|
21
|
Valentini V, Silvestri V, Marraffa F, Greco G, Bucalo A, Grassi S, Gagliardi A, Mazzotta A, Ottini L, Richetta AG. Gut microbiome profile in psoriatic patients treated and untreated with biologic therapy. J Dermatol 2021; 48:786-793. [PMID: 33511673 DOI: 10.1111/1346-8138.15680] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 10/09/2020] [Indexed: 01/06/2023]
Abstract
There are increasing data about the role of the gut microbiome in various autoimmune diseases, including psoriasis, a chronic inflammatory and immune-mediated disease. Current treatment strategies in psoriasis include immunomodulating biologic agents. A variable response to this type of therapy has been reported in psoriatic patients. A possible effect of biologic therapy on the gut microbiome composition has been suggested, but data are still limited. The aim of this study was to compare the gut microbiome composition between psoriatic patients treated and untreated with biologic drugs in order to identify differences which may highlight the potential impact of the treatment on the gut microbiome. 16S rRNA sequencing and bioinformatic analyses were performed on the fecal samples of 30 psoriatic patients with similar clinicopathological features, 10 of whom were undergoing biologic therapy and 20 not receiving systemic therapy. Alpha and beta diversity significantly differed between the two groups of patients. A reduced bacterial biodiversity in the group of treated patients compared with the group of untreated patients was observed. Differential relative abundances of key gut microbial communities, including Akkermansia muciniphila and Bacteroides plebeius, were identified between the two groups of patients. This study showed that biologic therapy may have an impact on the composition of the gut microbiome of psoriatic patients. Gut microbiome composition could be used as an indicator of response to therapy and the modulation of the microbial composition could help to restore the intestinal symbiosis in psoriatic patients.
Collapse
Affiliation(s)
- Virginia Valentini
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | | | - Federica Marraffa
- Unit of Dermatology, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy
| | - Giulia Greco
- Unit of Dermatology, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy
| | - Agostino Bucalo
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Sara Grassi
- Unit of Dermatology, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy
| | - Antonella Gagliardi
- Section of Microbiology, Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | | | - Laura Ottini
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Antonio Giovanni Richetta
- Unit of Dermatology, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
22
|
Fused Omics Data Models Reveal Gut Microbiome Signatures Specific of Inactive Stage of Juvenile Idiopathic Arthritis in Pediatric Patients. Microorganisms 2020; 8:microorganisms8101540. [PMID: 33036309 PMCID: PMC7650812 DOI: 10.3390/microorganisms8101540] [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: 09/14/2020] [Revised: 10/01/2020] [Accepted: 10/02/2020] [Indexed: 01/15/2023] Open
Abstract
Juvenile idiopathic arthritis (JIA) is the most common rheumatic disease in children. Herein, we evaluated the relationship between the gut microbiome (GM) and disease phenotype by an integrated omics fused approach. In a multicenter, observational cohort study, stools from Italian JIA patients were collected at baseline, active, and inactive disease stages, and their GM compared to healthy controls (CTRLs). The microbiota metabolome was analyzed to detect volatile- and non-volatile organic compounds (VOCs); the data were fused with operational taxonomic units (OTUs) from 16S RNA targeted-metagenomics and classified by chemometric models. Non-VOCs did not characterize JIA patients nor JIA activity stages compared to CTRLs. The core of VOCs, (Ethanol, Methyl-isobutyl-ketone, 2,6-Dimethyl-4-heptanone and Phenol) characterized patients at baseline and inactive disease stages, while the OTUs represented by Ruminococcaceae, Lachnospiraceae and Clostridiacea discriminated between JIA inactive stage and CTRLs. No differences were highlighted amongst JIA activity stages. Finally, the fused data discriminated inactive and baseline stages versus CTRLs, based on the contribution of the invariant core of VOCs while Ruminococcaceae concurred for the inactive stage versus CTRLs comparison. In conclusion, the GM signatures enabled to distinguish the inactive disease stage from CTRLs.
Collapse
|
23
|
The Echo of Pulmonary Tuberculosis: Mechanisms of Clinical Symptoms and Other Disease-Induced Systemic Complications. Clin Microbiol Rev 2020; 33:33/4/e00036-20. [PMID: 32611585 DOI: 10.1128/cmr.00036-20] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Clinical symptoms of active tuberculosis (TB) can range from a simple cough to more severe reactions, such as irreversible lung damage and, eventually, death, depending on disease progression. In addition to its clinical presentation, TB has been associated with several other disease-induced systemic complications, such as hyponatremia and glucose intolerance. Here, we provide an overview of the known, although ill-described, underlying biochemical mechanisms responsible for the clinical and systemic presentations associated with this disease and discuss novel hypotheses recently generated by various omics technologies. This summative update can assist clinicians to improve the tentative diagnosis of TB based on a patient's clinical presentation and aid in the development of improved treatment protocols specifically aimed at restoring the disease-induced imbalance for overall homeostasis while simultaneously eradicating the pathogen. Furthermore, future applications of this knowledge could be applied to personalized diagnostic and therapeutic options, bettering the treatment outcome and quality of life of TB patients.
Collapse
|
24
|
The risk of rheumatoid arthritis among patients with inflammatory bowel disease: a systematic review and meta-analysis. BMC Gastroenterol 2020; 20:192. [PMID: 32552882 PMCID: PMC7301504 DOI: 10.1186/s12876-020-01339-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 06/08/2020] [Indexed: 02/06/2023] Open
Abstract
Background Studies have suggested that patients with inflammatory bowel disease (IBD) have an increased risk of rheumatoid arthritis (RA). However, the available data on this association are inconsistent. This meta-analysis aimed to determine the association between IBD and the risk of RA. Methods Observational studies investigating the RA risk among patients with IBD (Crohn disease (CD) and/or ulcerative colitis (UC)) were searched in PubMed, Embase, and Web of Science from the date of inception to December 2019. The methodological quality of the included studies was assessed using the Newcastle-Ottawa Scale. Relative risks (RRs) and corresponding 95% confidential intervals (CIs) were pooled with a random-effects model. Heterogeneity was evaluated using I2 statistics while publication bias was determined using Begg’s and Egger’s tests. Subgroup and sensitivity analyses were performed. Results A total of three cohort studies, three cross-sectional studies, and two case-control studies were included in the meta-analyses. Compared to the non-IBD control or general population, there was a significantly higher risk of RA among patients with IBD (RR = 2.59; 95% CI: 1.93–3.48). Moreover, both CD (RR = 3.14; 95% CI: 2.46–4.01) and UC (RR = 2.29; 95% CI: 1.76–2.97) were associated with a significantly increased risk of RA. However, heterogeneity was substantial across studies and the subgroup analyses failed to identify the potential source of heterogeneity. Conclusions Patients with IBD have a greater risk of developing RA. Rheumatologists should be consulted when patients with IBD present with undifferentiated joint complaints. However, more prospective cohort studies are needed to validate these results.
Collapse
|
25
|
Gioia C, Lucchino B, Tarsitano MG, Iannuccelli C, Di Franco M. Dietary Habits and Nutrition in Rheumatoid Arthritis: Can Diet Influence Disease Development and Clinical Manifestations? Nutrients 2020; 12:nu12051456. [PMID: 32443535 PMCID: PMC7284442 DOI: 10.3390/nu12051456] [Citation(s) in RCA: 134] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/14/2020] [Accepted: 05/14/2020] [Indexed: 12/14/2022] Open
Abstract
Rheumatoid arthritis (RA) is a systemic, autoimmune disease characterized by joint involvement, with progressive cartilage and bone destruction. Genetic and environmental factors determine RA susceptibility. In recent years, an increasing number of studies suggested that diet has a central role in disease risk and progression. Several nutrients, such as polyunsaturated fatty acids, present anti-inflammatory and antioxidant properties, featuring a protective role for RA development, while others such as red meat and salt have a harmful effect. Gut microbiota alteration and body composition modifications are indirect mechanisms of how diet influences RA onset and progression. Possible protective effects of some dietary patterns and supplements, such as the Mediterranean Diet (MD), vitamin D and probiotics, could be a possible future adjunctive therapy to standard RA treatment. Therefore, a healthy lifestyle and nutrition have to be encouraged in patients with RA.
Collapse
Affiliation(s)
- Chiara Gioia
- Dipartimento di Scienze Cliniche, Internistiche, Anestesiologiche e Cardiovascolari-Reumatologia, Sapienza University of Rome, 00161 Roma, Italy; (C.G.); (C.I.); (M.D.F.)
| | - Bruno Lucchino
- Dipartimento di Scienze Cliniche, Internistiche, Anestesiologiche e Cardiovascolari-Reumatologia, Sapienza University of Rome, 00161 Roma, Italy; (C.G.); (C.I.); (M.D.F.)
- Correspondence: ; Tel.: +39-06-4997-4635
| | | | - Cristina Iannuccelli
- Dipartimento di Scienze Cliniche, Internistiche, Anestesiologiche e Cardiovascolari-Reumatologia, Sapienza University of Rome, 00161 Roma, Italy; (C.G.); (C.I.); (M.D.F.)
| | - Manuela Di Franco
- Dipartimento di Scienze Cliniche, Internistiche, Anestesiologiche e Cardiovascolari-Reumatologia, Sapienza University of Rome, 00161 Roma, Italy; (C.G.); (C.I.); (M.D.F.)
| |
Collapse
|
26
|
Pei J, Li F, Xie Y, Liu J, Yu T, Feng X. Microbial and metabolomic analysis of gingival crevicular fluid in general chronic periodontitis patients: lessons for a predictive, preventive, and personalized medical approach. EPMA J 2020; 11:197-215. [PMID: 32547651 PMCID: PMC7272536 DOI: 10.1007/s13167-020-00202-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 03/10/2020] [Indexed: 12/14/2022]
Abstract
Objectives General chronic periodontitis (GCP) is a bacterial inflammatory disease with complex pathology. Despite extensive studies published on the variation in the oral microbiota and metabolic profiles of GCP patients, information is lacking regarding the correlation between host-bacterial interactions and biochemical metabolism. This study aimed to analyze the oral microbiome, the oral metabolome, and the link between them and to identify potential molecules as useful biomarkers for predictive, preventive, and personalized medicine (PPPM) in GCP. Methods In this study, gingival crevicular fluid (GCF) samples were collected from patients with GCP (n = 30) and healthy controls (n = 28). The abundance of oral microbiota constituents was obtained by Illumina sequencing, and the relative level of metabolites was measured by gas chromatography-mass spectrometry. Full-mouth probing depth, clinical attachment loss, and bleeding on probing were recorded as indices of periodontal disease. Results The relative abundances of 7 phyla and 82 genera differed significantly between the GCP and healthy groups. Seventeen differential metabolites involved in different metabolism pathways were selected based on variable influence on projection values (VIP > 1) and P values (P < 0.05). Through Spearman's correlation analysis, microorganisms, metabolites in GCF, and clinical data together showed a clear trend, and clinical data regarding periodontitis can be reflected in the shift of the oral microbial community and the change in metabolites in GCF. A combination of citramalic acid and N-carbamylglutamate yielded satisfactory accuracy (AUC = 0.876) for the predictive diagnosis of GCP. Conclusions Dysbiosis in the polymicrobial community structure and changes in metabolism could be mechanisms underlying periodontitis. The differential microorganisms and metabolites in GCF between periodontitis patients and healthy individuals are possibly biomarkers, pointing to a potential strategy for the prediction, diagnosis, prognosis, and management of personalized periodontal therapy.
Collapse
Affiliation(s)
- Jun Pei
- Department of Preventive Dentistry, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200000 China.,National Clinical Research Center for Oral Diseases, Shanghai, 200000 China.,Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200000 China
| | - Fei Li
- Department of Preventive Dentistry, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200000 China.,National Clinical Research Center for Oral Diseases, Shanghai, 200000 China.,Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200000 China
| | - Youhua Xie
- Key Lab of Medical Molecular Virology, School of Basic Medical Sciences, Fudan University, Shanghai, 200000 China
| | - Jing Liu
- Key Lab of Medical Molecular Virology, School of Basic Medical Sciences, Fudan University, Shanghai, 200000 China
| | - Tian Yu
- Department of Preventive Dentistry, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200000 China.,National Clinical Research Center for Oral Diseases, Shanghai, 200000 China.,Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200000 China
| | - Xiping Feng
- Department of Preventive Dentistry, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200000 China.,National Clinical Research Center for Oral Diseases, Shanghai, 200000 China.,Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200000 China
| |
Collapse
|
27
|
Sanchez-Rodriguez E, Egea-Zorrilla A, Plaza-Díaz J, Aragón-Vela J, Muñoz-Quezada S, Tercedor-Sánchez L, Abadia-Molina F. The Gut Microbiota and Its Implication in the Development of Atherosclerosis and Related Cardiovascular Diseases. Nutrients 2020; 12:605. [PMID: 32110880 PMCID: PMC7146472 DOI: 10.3390/nu12030605] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Revised: 02/19/2020] [Accepted: 02/21/2020] [Indexed: 12/23/2022] Open
Abstract
The importance of gut microbiota in health and disease is being highlighted by numerous research groups worldwide. Atherosclerosis, the leading cause of heart disease and stroke, is responsible for about 50% of all cardiovascular deaths. Recently, gut dysbiosis has been identified as a remarkable factor to be considered in the pathogenesis of cardiovascular diseases (CVDs). In this review, we briefly discuss how external factors such as dietary and physical activity habits influence host-microbiota and atherogenesis, the potential mechanisms of the influence of gut microbiota in host blood pressure and the alterations in the prevalence of those bacterial genera affecting vascular tone and the development of hypertension. We will also be examining the microbiota as a therapeutic target in the prevention of CVDs and the beneficial mechanisms of probiotic administration related to cardiovascular risks. All these new insights might lead to novel analysis and CVD therapeutics based on the microbiota.
Collapse
Affiliation(s)
- Estefania Sanchez-Rodriguez
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain
- Institute of Nutrition and Food Technology “José Mataix”, Center of Biomedical Research, University of Granada, Avda. del Conocimiento s/n., 18016 Armilla, Granada, Spain;
| | - Alejandro Egea-Zorrilla
- Institute of Nutrition and Food Technology “José Mataix”, Center of Biomedical Research, University of Granada, Avda. del Conocimiento s/n., 18016 Armilla, Granada, Spain;
| | - Julio Plaza-Díaz
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain
- Institute of Nutrition and Food Technology “José Mataix”, Center of Biomedical Research, University of Granada, Avda. del Conocimiento s/n., 18016 Armilla, Granada, Spain;
- Instituto de Investigación Biosanitaria IBS.GRANADA, Complejo Hospitalario Universitario de Granada, 18014 Granada, Spain
| | - Jerónimo Aragón-Vela
- Department of Nutrition, Exercise and Sports (NEXS), Section of Integrative Physiology, University of Copenhagen, Nørre Allé 51, DK-2200 Copenhagen, Denmark;
| | - Sergio Muñoz-Quezada
- Departamento de Farmacia, Facultad de Química, Pontificia Universidad Católica de Chile, Santiago 6094411, Chile;
- National Agency for Medicines (ANAMED), Public Health Institute, Santiago 7780050, Chile
| | | | - Francisco Abadia-Molina
- Institute of Nutrition and Food Technology “José Mataix”, Center of Biomedical Research, University of Granada, Avda. del Conocimiento s/n., 18016 Armilla, Granada, Spain;
- Department of Cell Biology, School of Sciences, University of Granada, 18071 Granada, Spain
| |
Collapse
|
28
|
Białowąs K, Radwan-Oczko M, Duś-Ilnicka I, Korman L, Świerkot J. Periodontal disease and influence of periodontal treatment on disease activity in patients with rheumatoid arthritis and spondyloarthritis. Rheumatol Int 2019; 40:455-463. [PMID: 31701185 DOI: 10.1007/s00296-019-04460-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 10/03/2019] [Indexed: 01/18/2023]
Abstract
The aim of this study was to assess the prevalence of periodontal disease and the effect of periodontal treatment in patients with rheumatoid arthritis (RA) and spondyloarthritis (SpA). Forty-four RA patients, thirty SpA patients and thirty-nine healthy volunteers were recruited to the study. Periodontal examination included the approximal plaque index (API), bleeding on probing (BoP), probing depth (PD) and number of teeth. Samples from the deepest periodontal pockets were taken for the detection of Porphyromonas gingivalis DNA with the use of the polymerase chain reaction. All subjects with periodontitis, who completed the study, received periodontal treatment consisting of scaling/root planing and oral hygiene instructions. Disease activity scores, clinical and laboratory parameters were assessed before and 4-6 weeks after periodontal treatment. No significant difference in the prevalence of periodontal disease and the presence of P. gingivalis DNA were found in RA and SpA patients compared to healthy controls. Significantly higher API (80% vs 63%; p = 0.01) and a lower number of teeth (20 vs 25, p = 0.001) were found in RA patients. BoP was significantly elevated in SpA patients (51% vs 33%, p = 0.02). Disease activity measured by the DAS28(CRP) was significantly reduced in RA patients after periodontal treatment (p = 0.002). Clinical and biochemical parameters were not improved in SpA patients. Nonsurgical periodontal treatment had an impact on the decrease in RA activity. Periodontal examination is necessary in patients with RA to detect and treat periodontitis at an early stage.
Collapse
Affiliation(s)
- Katarzyna Białowąs
- Department of Rheumatology and Internal Medicine, Wroclaw Medical University, Ulica Borowska 213, 50-556, Wroclaw, Poland.
| | - Małgorzata Radwan-Oczko
- Department of Oral Pathology, Wroclaw Medical University, Ulica Krakowska 26, 50-425, Wroclaw, Poland
| | - Irena Duś-Ilnicka
- Department of Oral Pathology, Wroclaw Medical University, Ulica Krakowska 26, 50-425, Wroclaw, Poland
| | - Lucyna Korman
- Department of Rheumatology and Internal Medicine, Wroclaw Medical University, Ulica Borowska 213, 50-556, Wroclaw, Poland
| | - Jerzy Świerkot
- Department of Rheumatology and Internal Medicine, Wroclaw Medical University, Ulica Borowska 213, 50-556, Wroclaw, Poland
| |
Collapse
|
29
|
Linkage of Periodontitis and Rheumatoid Arthritis: Current Evidence and Potential Biological Interactions. Int J Mol Sci 2019; 20:ijms20184541. [PMID: 31540277 PMCID: PMC6769683 DOI: 10.3390/ijms20184541] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 08/07/2019] [Accepted: 08/20/2019] [Indexed: 02/06/2023] Open
Abstract
The association between rheumatoid arthritis (RA) and periodontal disease (PD) has been the focus of numerous investigations driven by their common pathological features. RA is an autoimmune disease characterized by chronic inflammation, the production of anti-citrullinated proteins antibodies (ACPA) leading to synovial joint inflammation and destruction. PD is a chronic inflammatory condition associated with a dysbiotic microbial biofilm affecting the supporting tissues around the teeth leading to the destruction of mineralized and non-mineralized connective tissues. Chronic inflammation associated with both RA and PD is similar in the predominant adaptive immune phenotype, in the imbalance between pro- and anti-inflammatory cytokines and in the role of smoking and genetic background as risk factors. Structural damage that occurs in consequence of chronic inflammation is the ultimate cause of loss of function and disability observed with the progression of RA and PD. Interestingly, the periodontal pathogen Porphyromonas gingivalis has been implicated in the generation of ACPA in RA patients, suggesting a direct biological intersection between PD and RA. However, more studies are warranted to confirm this link, elucidate potential mechanisms involved, and ascertain temporal associations between RA and PD. This review is mainly focused on recent clinical and translational research intends to discuss and provide an overview of the relationship between RA and PD, exploring the similarities in the immune-pathological aspects and the possible mechanisms linking the development and progression of both diseases. In addition, the current available treatments targeting both RA and PD were revised.
Collapse
|
30
|
Hidalgo-Cantabrana C, Gómez J, Delgado S, Requena-López S, Queiro-Silva R, Margolles A, Coto E, Sánchez B, Coto-Segura P. Gut microbiota dysbiosis in a cohort of patients with psoriasis. Br J Dermatol 2019; 181:1287-1295. [PMID: 30920647 DOI: 10.1111/bjd.17931] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/18/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND There is increasing evidence of the key role that the gut microbiota plays in inflammatory diseases. OBJECTIVES To identify differences in the faecal microbial composition of patients with psoriasis compared with healthy individuals in order to unravel the microbiota profiling in this autoimmune disease. METHODS 16S rRNA gene sequencing and bioinformatic analyses were performed with the total DNA extracted from the faecal microbiota of 19 patients with psoriasis and 20 healthy individuals from the same geographic location. RESULTS Gut microbiota composition of patients with psoriasis displayed a lower diversity and different relative abundance of certain bacterial taxa compared with healthy individuals. CONCLUSIONS The gut microbiota profile of patients with psoriasis displayed a clear dysbiosis that can be targeted for microbiome-based therapeutic approaches. What's already known about this topic? Psoriasis is a chronic inflammatory immune-mediated skin disease, the aetiology of which remains unclear. The human microbiota is a complex microbial community that inhabits our body and has been related with the maintenance of a healthy status. Several studies have focused on the skin microbiome and its connection with psoriasis although less attention has been focused on the potential role of the gut microbiota in psoriatic disease. What does this study add? This study unravels the gut microbiome dysbiosis present in a cohort of patients with psoriasis, compared with a healthy control group from the same geographical location. This study shows a lower bacterial diversity and different relative abundance of certain bacterial taxa in patients with psoriasis. We gain knowledge and insight into the microbiome alterations in psoriatic disease, opening new avenues for therapeutic approaches to reshape the human microbiome towards a healthy status.
Collapse
Affiliation(s)
- C Hidalgo-Cantabrana
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias-Consejo Superior de Investigaciones Científicas (IPLA-CSIC), Paseo Río Linares s/n 33300 Villaviciosa, Asturias, Spain
| | - J Gómez
- Molecular Genetics Laboratory, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - S Delgado
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias-Consejo Superior de Investigaciones Científicas (IPLA-CSIC), Paseo Río Linares s/n 33300 Villaviciosa, Asturias, Spain
| | - S Requena-López
- Dermatology and, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - R Queiro-Silva
- Department of Rheumatology, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - A Margolles
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias-Consejo Superior de Investigaciones Científicas (IPLA-CSIC), Paseo Río Linares s/n 33300 Villaviciosa, Asturias, Spain
| | - E Coto
- Molecular Genetics Laboratory, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - B Sánchez
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias-Consejo Superior de Investigaciones Científicas (IPLA-CSIC), Paseo Río Linares s/n 33300 Villaviciosa, Asturias, Spain
| | - P Coto-Segura
- Department of Dermatology, Hospital Vital Álvarez-Buylla, Mieres, Spain
| |
Collapse
|
31
|
Yamashita T, Kawada-Matsuo M, Katsumata T, Watanabe A, Oogai Y, Nishitani Y, Miyawaki S, Komatsuzawa H. Antibacterial activity of disodium succinoyl glycyrrhetinate, a derivative of glycyrrhetinic acid against Streptococcus mutans. Microbiol Immunol 2019; 63:251-260. [PMID: 31166029 DOI: 10.1111/1348-0421.12717] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 05/09/2019] [Accepted: 05/23/2019] [Indexed: 11/28/2022]
Abstract
Streptococcus mutans is a cariogenic bacterium that localizes in the oral cavity. Glycyrrhetinic acid (GRA) is a major component of licorice extract. GRA and several derivatives, including disodium succinoyl glycyrrhetinate (GR-SU), are known to have anti-inflammatory effects in humans. In this study, the antimicrobial effect of GRA and its derivatives against the S. mutans UA159 strain were investigated. Minimum inhibitory concentrations (MICs) of GRA and GR-SU showed antibacterial activity against the S. mutans strain, whereas other tested derivatives did not. Because GR-SU is more soluble than GRA, GR-SU was used for further experiments. The antibacterial activity of GR-SU against 100 S. mutans strains was evaluated and it was found that all strains are susceptible to GR-SU, with MIC values below 256 µg/mL. A cell viability assay showed that GR-SU has a bacteriostatic effect on S. mutans cells. As to growth kinetics, sub-MICs of GR-SU inhibited growth. The effect of GR-SU on S. mutans virulence was then investigated. GR-SU at sub-MICs suppresses biofilm formation. Additionally, GR-SU greatly suppresses the pH drop caused by the addition of glucose and glucose-induced expression of the genes responsible for acid production (ldh and pykF) and tolerance (aguD and atpD). Additionally, expression of enolase, which is responsible for the carbohydrate phosphotransferase system, was not increased in the presence of GR-SU, indicating that GR-SU suppresses incorporation of sugars into S. mutans. In conclusion, GR-SU has antibacterial activity against S. mutans and also decreases S. mutans virulence.
Collapse
Affiliation(s)
- Takahito Yamashita
- Department of Oral Microbiology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Miki Kawada-Matsuo
- Department of Oral Microbiology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Tamaki Katsumata
- Department of Restorative Dentistry and Endodontology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Atsuko Watanabe
- Department of Orthodontics, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Yuichi Oogai
- Department of Oral Microbiology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Yoshihiro Nishitani
- Department of Restorative Dentistry and Endodontology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Shouichi Miyawaki
- Department of Orthodontics, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Hitoshi Komatsuzawa
- Department of Bacteriology, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| |
Collapse
|
32
|
Wang Z, Qi Q. Gut microbial metabolites associated with HIV infection. Future Virol 2019; 14:335-347. [PMID: 31263508 PMCID: PMC6595475 DOI: 10.2217/fvl-2019-0002] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 04/25/2019] [Indexed: 02/06/2023]
Abstract
HIV infection has been associated with alterations in gut microbiota and related microbial metabolite production. However, the mechanisms of how these functional microbial metabolites may affect HIV immunopathogenesis and comorbidities, such as cardiovascular disease and other metabolic diseases, remain largely unknown. Here we review the current understanding of gut microbiota and related metabolites in the context of HIV infection. We focus on several bacteria-produced metabolites, including tryptophan catabolites, short-chain fatty acids and trimethylamine-N-oxide (TMAO), and discuss their implications in HIV infection and comorbidities. We also prospect future studies using integrative multiomics approaches to better understand host-microbiota-metabolites interactions in HIV infection, and facilitate integrative medicine utilizing the microbiota in HIV infection.
Collapse
Affiliation(s)
- Zheng Wang
- Department of Epidemiology & Population Health, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Qibin Qi
- Department of Epidemiology & Population Health, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
33
|
Volokh O, Klimenko N, Berezhnaya Y, Tyakht A, Nesterova P, Popenko A, Alexeev D. Human Gut Microbiome Response Induced by Fermented Dairy Product Intake in Healthy Volunteers. Nutrients 2019; 11:nu11030547. [PMID: 30836671 PMCID: PMC6470569 DOI: 10.3390/nu11030547] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 02/18/2019] [Accepted: 02/26/2019] [Indexed: 02/07/2023] Open
Abstract
Accumulated data suggests that the gut microbiome can rapidly respond to changes in diet. Consumption of fermented dairy products (FDP) fortified with probiotic microbes may be associated with positive impact on human health. However, the extent and details of the possible impact of FDP consumption on gut community structure tends to vary across individuals. We used microbiome analysis to characterize changes in gut microbiota composition after 30 days of oral intake of a yoghurt fortified with Bifidobacterium animalis subsp. lactis BB-12. 16S rRNA gene sequencing was used to assess the gut microbial composition before and after FDP consumption in healthy adults (n = 150). Paired comparison of gut microbial content demonstrated an increase in presence of potentially beneficial bacteria, particularly, Bifidobacterium genus, as well as Adlercreutzia equolifaciens and Slackia isoflavoniconvertens. At a functional level, an increased capacity to metabolize lactose and synthesize amino acids was observed accompanied by a lowered potential for synthesis of lipopolysaccharides. Cluster analysis revealed that study volunteers segregated into two groups with post-intervention microbiota response that was dependent on the baseline microbial community structure.
Collapse
Affiliation(s)
- Olesya Volokh
- PepsiCo R&D, Inc., Leningradsky prospekt, 72-4, 125315 Moscow, Russia.
| | - Natalia Klimenko
- Knomics LLC, Skolkovo Innovation Center, Bolshoy bulvar street 42-1, 143026 Moscow, Russia.
- Institute of Gene Biology of Russian Academy of Science, Group of Molecular Organization of Genome, 34/5 Vavilova Str., 119334 Moscow, Russia.
| | - Yulia Berezhnaya
- PepsiCo R&D, Inc., Leningradsky prospekt, 72-4, 125315 Moscow, Russia.
| | - Alexander Tyakht
- Knomics LLC, Skolkovo Innovation Center, Bolshoy bulvar street 42-1, 143026 Moscow, Russia.
- Institute of Gene Biology of Russian Academy of Science, Group of Molecular Organization of Genome, 34/5 Vavilova Str., 119334 Moscow, Russia.
- ITMO University, Computer Technology Department, Kronverkskiy pr., 49, 197101 St. Petersburg, Russia.
| | - Polina Nesterova
- PepsiCo R&D, Inc., Leningradsky prospekt, 72-4, 125315 Moscow, Russia.
| | - Anna Popenko
- Knomics LLC, Skolkovo Innovation Center, Bolshoy bulvar street 42-1, 143026 Moscow, Russia.
| | - Dmitry Alexeev
- Knomics LLC, Skolkovo Innovation Center, Bolshoy bulvar street 42-1, 143026 Moscow, Russia.
- ITMO University, Computer Technology Department, Kronverkskiy pr., 49, 197101 St. Petersburg, Russia.
- Atlas Biomed Group, 92 Albert Embankment, Lambeth, London SE1 7TT, UK.
| |
Collapse
|
34
|
Li W, Xu X, Wen H, Wang Z, Ding C, Liu X, Gao Y, Su H, Zhang J, Han Y, Xia Y, Wang X, Gu H, Yao X. Inverse Association Between the Skin and Oral Microbiota in Atopic Dermatitis. J Invest Dermatol 2019; 139:1779-1787.e12. [PMID: 30802424 DOI: 10.1016/j.jid.2019.02.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 02/01/2019] [Accepted: 02/04/2019] [Indexed: 12/21/2022]
Abstract
Previous studies have shown independently that the skin and gut microbiota are closely associated with atopic dermatitis (AD); however, the microbiota across different habitats of AD patients as an integrated community has not been characterized. In the present study, we comparatively analyzed the structure and function of the microbial communities in the skin, oral cavity, and gut of 172 AD patients and 120 healthy controls through 16S ribosomal RNA gene amplicon sequencing. The skin and oral cavity, but not the gut, of AD patients demonstrated differential reduction in the microbial diversity, and these were distinctly correlated with disease severity. Different degrees of shifts in the community structure were found among different habitats, and the lineage distance between the skin and oral microbiota of AD patients was closer than that observed in the controls. The different habitats of AD patients exhibited site-specific alterations at the genus level, and many oral-specific microbes of AD showed opposing directions of enrichment in the skin and oral cavity. Most interestingly, an inverse association in the functional pathways was found between the skin and oral microbiota of AD patients. Additionally, the alterations of the microbiota in different body sites of AD patients were differentially affected by age.
Collapse
Affiliation(s)
- Wei Li
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, People's Republic of China; Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Xiaoqiang Xu
- Aimigene Institute, Shenzhen, People's Republic of China
| | - He Wen
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, People's Republic of China
| | - Zhifeng Wang
- Aimigene Institute, Shenzhen, People's Republic of China
| | - Chao Ding
- Department of General Surgery, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, People's Republic of China; Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, People's Republic of China
| | - Xiaochun Liu
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, People's Republic of China
| | - Yingxia Gao
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, People's Republic of China
| | - Huichun Su
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, People's Republic of China
| | - Jingxi Zhang
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, People's Republic of China
| | - Yue Han
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, People's Republic of China
| | - Yan Xia
- Aimigene Institute, Shenzhen, People's Republic of China
| | - Xiaokai Wang
- Aimigene Institute, Shenzhen, People's Republic of China
| | - Heng Gu
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, People's Republic of China.
| | - Xu Yao
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, People's Republic of China.
| |
Collapse
|
35
|
De Filippo C, Di Paola M, Giani T, Tirelli F, Cimaz R. Gut microbiota in children and altered profiles in juvenile idiopathic arthritis. J Autoimmun 2019; 98:1-12. [PMID: 30638708 DOI: 10.1016/j.jaut.2019.01.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 12/19/2018] [Accepted: 01/02/2019] [Indexed: 12/18/2022]
Abstract
Microbial diversity plays a key role in the maintenance of intestinal homeostasis and in the development of the immune system in the gut mucosa. Maybe one of the most important function of our gut microbiota is the immune system education, in particular the discrimination of friends from foes that occurs during childhood. In addition to bacterial antigens, several metabolites of microbial origin have a crucial role in training of the immune system, such as Short Chain Fatty Acids (SCFAs). There are many evidences on the role of the gut microbiota in rheumatic diseases, in particular modifications of microbiota composition causing dysbiosis that, in turn, can induce gut permeability, and thus immunological imbalance and trigger inflammation. In particular, immune cells can reach extra-intestinal sites, such as joints and trigger local inflammation. Childhood is a crucial period of life for development and evolution of the gut microbiota, especially for the acquisition of fundamental functions such as immunotolerance of commensal microorganisms. For this reason, gut dysbiosis is gaining interest as a potential pathogenetic factor for Juvenile Idiopathic Arthritis (JIA). Here we summarized the studies conducted on JIA patients in which a pro-arthritogenic microbial profiles has been observed; this, together with a depletion of microbial biodiversity, clearly distinguish patients' from healthy subjects' microbiota. Further studies are however needed to better clarify the role of microbiota in JIA pathogenesis.
Collapse
Affiliation(s)
- Carlotta De Filippo
- Institute of Biology and Agrarian Biotechnology (IBBA), National Research Council (CNR), Via Moruzzi 1, 56124 Pisa, Italy
| | - Monica Di Paola
- Department of Biology, University of Florence, Via Madonna del Piano 6, 50019 Sesto Fiorentino, Florence, Italy
| | - Teresa Giani
- Rheumatology Unit, Anna Meyer Children's Hospital, University of Florence, Viale G. Pieraccini 24, 50139, Florence, Italy; Department of Medica Biotechnologies, University of Siena, Viale Mario Bracci, 16 53100, Siena, Italy
| | - Francesca Tirelli
- Rheumatology Unit, Anna Meyer Children's Hospital, University of Florence, Viale G. Pieraccini 24, 50139, Florence, Italy
| | - Rolando Cimaz
- Rheumatology Unit, Anna Meyer Children's Hospital, University of Florence, Viale G. Pieraccini 24, 50139, Florence, Italy; Department of Neuroscience, Psychology, Drug Research and Child Health, Meyer Children's Hospital, University of Florence, Viale G. Pieraccini 6, 50139, Florence, Italy.
| |
Collapse
|
36
|
Lei Y, Zhang K, Guo M, Li G, Li C, Li B, Yang Y, Chen Y, Wang X. Exploring the Spatial-Temporal Microbiota of Compound Stomachs in a Pre-weaned Goat Model. Front Microbiol 2018; 9:1846. [PMID: 30158908 PMCID: PMC6104157 DOI: 10.3389/fmicb.2018.01846] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 07/24/2018] [Indexed: 12/16/2022] Open
Abstract
Ruminant animals possess a characteristic four-compartment stomach (rumen, reticulum, omasum, and abomasum) that is specialized for pre-intestinal digestion of plant materials. Of these four compartments, the rumen is the largest. The rumen's diverse microbial community has been well studied. However, the current understanding of microbial profiles in the reticulum, omasum and abomasum are lacking. In the present study, fluid samples from the reticulum, omasum, and abomasum of goats at 3, 7, 14, 21, 28, 42, and 56 days after birth, as well as the negative controls (NC) used for microbial DNA extraction, were subjected to 16S rRNA sequencing. By filtering operational taxonomic units (OTUs) in NC, distinct temporal distributions of microbes were observed in the different compartments, we showed that the OTUs in control samples had a large effect to the samples with low microbial density. In addition, Proteobacteria gradually decreased with age from days 3 to 56 in all three compartments, and the relative abundance of Bacteroidetes increased from 24.15% (Day 3) to 52.03% (Day 56) in abomasum. Network analysis revealed that Prevotellaceae_UGG-03 and Rikenellaceae_RC9 were positively correlated with Prevotella_1, lending support to the well understood fact that cellulose is well digested in compound stomachs prior to the rumen. Pathway analysis revealed that gene expression in abomasum at Day 3 were primarily related to Glycolysis/Gluconeogenesis and Pyruvate metabolism, suggesting that colostrum digestion is the dominant function of the abomasum at an early age. These findings combined with other recent rumen microbiota data show that the microbiome landscape represents three distinct stages in ruminant stomachs. The first stage is to gain access to external microorganisms at Day 0-14, the secondary stage is for microbial transition at Day 14-28, and the third stage is for exogenous and endogenous microbial colonization beyond Day 28 of age. Our results provide insight into microbiota dynamics in ruminant stomachs, and will facilitate efforts for the maintenance of gastrointestinal balance and intervention with starter diets in juvenile ruminants during early development.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Xiaolong Wang
- College of Animal Science and Technology, Northwest A&F University, Xianyang, China
| |
Collapse
|
37
|
Impact of obesity on autoimmune arthritis and its cardiovascular complications. Autoimmun Rev 2018; 17:821-835. [PMID: 29885537 PMCID: PMC9996646 DOI: 10.1016/j.autrev.2018.02.007] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 02/25/2018] [Indexed: 02/06/2023]
Abstract
Obesity can instigate and sustain a systemic low-grade inflammatory environment that can amplify autoimmune disorders and their associated comorbidities. Metabolic changes and inflammatory factors produced by the adipose tissue have been reported to aggravate autoimmunity and predispose the patient to cardiovascular disease (CVD) and metabolic comorbidities. Rheumatoid arthritis (RA) and psoriatic arthritis (PsA) are autoimmune arthritic diseases, often linked with altered body mass index (BMI). Severe joint inflammation and bone destruction have a debilitating impact on the patient's life; there is also a staggering risk of cardiovascular morbidity and mortality. Furthermore, these patients are at risk of developing metabolic symptoms, including insulin resistance resulting in type 2 diabetes mellitus (T2DM). In addition, arthritis severity, progression and response to therapy can be markedly affected by the patient's BMI. Hence, a complex integrative pathogenesis interconnects autoimmunity with metabolic and cardiovascular disorders. This review aims to shed light on the network that connects obesity with RA, PsA, systemic lupus erythematosus and Sjӧgren's syndrome. We have focused on clarifying the mechanism by which obesity affects different cell types, inflammatory factors and traditional therapies in these autoimmune disorders. We conclude that to further optimize arthritis therapy and to prevent CVD, it is imperative to uncover the intricate relation between obesity and arthritis pathology.
Collapse
|
38
|
Zhu CS, Grandhi R, Patterson TT, Nicholson SE. A Review of Traumatic Brain Injury and the Gut Microbiome: Insights into Novel Mechanisms of Secondary Brain Injury and Promising Targets for Neuroprotection. Brain Sci 2018; 8:brainsci8060113. [PMID: 29921825 PMCID: PMC6025245 DOI: 10.3390/brainsci8060113] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 06/15/2018] [Accepted: 06/17/2018] [Indexed: 12/16/2022] Open
Abstract
The gut microbiome and its role in health and disease have recently been major focus areas of research. In this review, we summarize the different ways in which the gut microbiome interacts with the rest of the body, with focus areas on its relationships with immunity, the brain, and injury. The gut–brain axis, a communication network linking together the central and enteric nervous systems, represents a key bidirectional pathway with feed-forward and feedback mechanisms. The gut microbiota has a central role in this pathway and is significantly altered following injury, leading to a pro-inflammatory state within the central nervous system (CNS). Herein, we examine traumatic brain injury (TBI) in relation to this axis and explore potential interventions, which may serve as targets for improving clinical outcomes and preventing secondary brain injury.
Collapse
Affiliation(s)
- Caroline S Zhu
- Division of Trauma and Emergency Surgery, Department of Surgery, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive (MC 7740), San Antonio, TX 78229, USA.
| | - Ramesh Grandhi
- Division of Trauma and Emergency Surgery, Department of Surgery, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive (MC 7740), San Antonio, TX 78229, USA.
- Department of Neurosurgery, The University of Texas Health Sciences Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA.
- Department of Neurosurgery, The University of Utah School of Medicine, Salt Lake City, UT 84132, USA.
| | - Thomas Tyler Patterson
- Division of Trauma and Emergency Surgery, Department of Surgery, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive (MC 7740), San Antonio, TX 78229, USA.
| | - Susannah E Nicholson
- Division of Trauma and Emergency Surgery, Department of Surgery, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive (MC 7740), San Antonio, TX 78229, USA.
| |
Collapse
|
39
|
Ursini F, Russo E, Pellino G, D'Angelo S, Chiaravalloti A, De Sarro G, Manfredini R, De Giorgio R. Metformin and Autoimmunity: A "New Deal" of an Old Drug. Front Immunol 2018; 9:1236. [PMID: 29915588 PMCID: PMC5994909 DOI: 10.3389/fimmu.2018.01236] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 05/17/2018] [Indexed: 12/12/2022] Open
Abstract
Metformin (dimethyl biguanide) is a synthetic derivative of guanidine, isolated from the extracts of Galega officinalis, a plant with a prominent antidiabetic effect. Since its discovery more than 50 years ago, metformin represents a worldwide milestone in treatment of patients with type 2 diabetes (T2D). Recent evidence in humans indicates novel pleiotropic actions of metformin which span from its consolidated role in T2D management up to various regulatory properties, including cardio- and nephro-protection, as well as antiproliferative, antifibrotic, and antioxidant effects. These findings, together with ground-breaking studies demonstrating its ability to prolong healthspan and lifespan in mice, provided the basis for defining metformin as a potential antiaging molecule. Moreover, emerging in vivo and in vitro evidence support the novel hypothesis that metformin can exhibit immune-modulatory features. Studies suggest that metformin interferes with key immunopathological mechanisms involved in systemic autoimmune diseases, such as the T helper 17/regulatory T cell balance, germinal centers formation, autoantibodies production, macrophage polarization, cytokine synthesis, neutrophil extracellular traps release, and bone or extracellular matrix remodeling. These effects may represent a powerful contributor to antiaging and anticancer properties exerted by metformin and, from another standpoint, may open the way to assess whether metformin can be a candidate molecule for clinical trials involving patients with immune-mediated diseases. In this article, we will review the available preclinical and clinical evidence regarding the effect of metformin on individual cells of the immune system, with emphasis on immunological mechanisms related to the development and maintenance of autoimmunity and its potential relevance in treatment of autoimmune diseases.
Collapse
Affiliation(s)
- Francesco Ursini
- Department of Health Sciences, University of Catanzaro "Magna Graecia", Catanzaro, Italy
| | - Emilio Russo
- Department of Health Sciences, University of Catanzaro "Magna Graecia", Catanzaro, Italy
| | - Gianluca Pellino
- Colorectal Unit, Hospital Universitario y Politécnico La Fe, Valencia, Spain.,Department of Medical, Surgical, Neurological, Metabolic and Ageing Sciences, Università della Campania "Luigi Vanvitelli", Naples, Italy
| | - Salvatore D'Angelo
- Rheumatology Institute of Lucania (IReL) - Rheumatology Department of Lucania, "San Carlo" Hospital of Potenza and "Madonna delle Grazie" Hospital of Matera, Potenza, Italy.,Basilicata Ricerca Biomedica (BRB) Foundation, Potenza, Italy
| | - Agostino Chiaravalloti
- Department of Biomedicine and Prevention, University Tor Vergata, Rome, Italy.,Department of Nuclear Medicine, IRCCS Neuromed, Pozzilli, Italy
| | | | - Roberto Manfredini
- Department of Medical Sciences, Clinica Medica Unit, University of Ferrara, Ferrara, Italy
| | - Roberto De Giorgio
- Department of Medical Sciences, Clinica Medica Unit, University of Ferrara, Ferrara, Italy
| |
Collapse
|
40
|
Luo Y, Blackledge WC. Microbiome-based mechanisms hypothesized to initiate obesity-associated rheumatoid arthritis. Obes Rev 2018; 19:786-797. [PMID: 29426061 DOI: 10.1111/obr.12671] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 12/11/2017] [Accepted: 12/18/2017] [Indexed: 02/05/2023]
Abstract
Worldwide, the growing obesity pandemic contributes to a range of chronic diseases. Recent epidemiological studies have suggested an association between obesity and the development of rheumatoid arthritis (RA), particularly among young women, whereby pro-inflammatory effects of adipokines provide one explanatory hypothesis. Yet, recent clinical and laboratory-based studies provide emerging evidence indicating microbiome involvement in RA initiation and development, including anti-citrullinated antibody formation and Th17 cell activation. Obesity and RA-associated microbiome alteration might provide a plausible link to address the impact of obesity to RA pathogenesis. The microbiome's influence on RA development - at mucosal as well as articular sites - and relevant pathophysiological mechanisms regarding obesity's association with RA are presented herein to discuss this hypothesis and aid understanding of obesity's role in RA development.
Collapse
Affiliation(s)
- Y Luo
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | | |
Collapse
|
41
|
Xun Z, Zhang Q, Xu T, Chen N, Chen F. Dysbiosis and Ecotypes of the Salivary Microbiome Associated With Inflammatory Bowel Diseases and the Assistance in Diagnosis of Diseases Using Oral Bacterial Profiles. Front Microbiol 2018; 9:1136. [PMID: 29899737 PMCID: PMC5988890 DOI: 10.3389/fmicb.2018.01136] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 05/14/2018] [Indexed: 12/16/2022] Open
Abstract
Inflammatory bowel diseases (IBDs) are chronic, idiopathic, relapsing disorders of unclear etiology affecting millions of people worldwide. Aberrant interactions between the human microbiota and immune system in genetically susceptible populations underlie IBD pathogenesis. Despite extensive studies examining the involvement of the gut microbiota in IBD using culture-independent techniques, information is lacking regarding other human microbiome components relevant to IBD. Since accumulated knowledge has underscored the role of the oral microbiota in various systemic diseases, we hypothesized that dissonant oral microbial structure, composition, and function, and different community ecotypes are associated with IBD; and we explored potentially available oral indicators for predicting diseases. We examined the 16S rRNA V3–V4 region of salivary bacterial DNA from 54 ulcerative colitis (UC), 13 Crohn’s disease (CD), and 25 healthy individuals using Illumina sequencing. Distinctive sample clusters were driven by disease or health based on principal coordinate analysis (PCoA) of both the Operational Taxonomic Unit profile and Kyoto Encyclopedia of Genes and Genomes pathways. Comparisons of taxa abundances revealed enrichment of Streptococcaceae (Streptococcus) and Enterobacteriaceae in UC and Veillonellaceae (Veillonella) in CD, accompanied by depletion of Lachnospiraceae and [Prevotella] in UC and Neisseriaceae (Neisseria) and Haemophilus in CD, most of which have been demonstrated to exhibit the same variation tendencies in the gut of IBD patients. IBD-related oral microorganisms were associated with white blood cells, reduced basic metabolic processes, and increased biosynthesis and transport of substances facilitating oxidative stress and virulence. Furthermore, UC and CD communities showed robust sub-ecotypes that were not demographic or severity-specific, suggesting their value for future applications in precision medicine. Additionally, indicator species analysis revealed several genera indicative of UC and CD, which were confirmed in a longitudinal cohort. Collectively, this study demonstrates evident salivary dysbiosis and different ecotypes in IBD communities and provides an option for identifying at-risk populations, not only enhancing our understanding of the IBD microbiome apart from the gut but also offering a clinically useful strategy to track IBD as saliva can be sampled conveniently and non-invasively.
Collapse
Affiliation(s)
- Zhe Xun
- Department of Preventive Dentistry, Peking University School and Hospital of Stomatology, Beijing, China
| | - Qian Zhang
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, China.,National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Tao Xu
- Department of Preventive Dentistry, Peking University School and Hospital of Stomatology, Beijing, China
| | - Ning Chen
- Department of Gastroenterology, Peking University People's Hospital, Beijing, China
| | - Feng Chen
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, China.,National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing, China
| |
Collapse
|
42
|
Chimenti MS, Perricone C, Novelli L, Caso F, Costa L, Bogdanos D, Conigliaro P, Triggianese P, Ciccacci C, Borgiani P, Perricone R. Interaction between microbiome and host genetics in psoriatic arthritis. Autoimmun Rev 2018; 17:276-283. [DOI: 10.1016/j.autrev.2018.01.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 11/06/2017] [Indexed: 12/21/2022]
|
43
|
Badsha H. Role of Diet in Influencing Rheumatoid Arthritis Disease Activity. Open Rheumatol J 2018; 12:19-28. [PMID: 29515679 PMCID: PMC5827298 DOI: 10.2174/1874312901812010019] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Revised: 01/06/2018] [Accepted: 01/21/2018] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Patients with Rheumatoid Arthritis (RA) frequently ask their doctors about which diets to follow, and even in the absence of advice from their physicians, many patients are undertaking various dietary interventions. DISCUSSION However, the role of dietary modifications in RA is not well understood. Several studies have tried to address these gaps in our understanding. Intestinal microbial modifications are being studied for the prevention and management of RA. Some benefits of vegan diet may be explained by antioxidant constituents, lactobacilli and fibre, and by potential changes in intestinal flora. Similarly, Mediterranean diet shows anti-inflammatory effects due to protective properties of omega-3 polyunsaturated fatty acids and vitamins, but also by influencing the gut microbiome. Gluten-free and elemental diets have been associated with some benefits in RA though the existing evidence is limited. Long-term intake of fish and other sources of long-chain polyunsaturated fatty acids are protective for development of RA. The benefits of fasting, anti-oxidant supplementation, flavanoids, and probiotics in RA are not clear. Vitamin D has been shown to influence autoimmunity and specifically decrease RA disease activity. The role of supplements such as fish oils and vitamin D should be explored in future trials to gain new insights in disease pathogenesis and develop RA-specific dietary recommendations. CONCLUSION Specifically more research is needed to explore the association of diet and the gut microbiome and how this can influence RA disease activity.
Collapse
Affiliation(s)
- Humeira Badsha
- Dr. Humeira Badsha Medical Center, Beach Park Plaza, Jumeira Road, Dubai, UAE
| |
Collapse
|
44
|
Bora S, Rindfleisch JA. The Elimination Diet. Integr Med (Encinitas) 2018. [DOI: 10.1016/b978-0-323-35868-2.00086-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
45
|
Yadav M, Verma MK, Chauhan NS. A review of metabolic potential of human gut microbiome in human nutrition. Arch Microbiol 2017; 200:203-217. [PMID: 29188341 DOI: 10.1007/s00203-017-1459-x] [Citation(s) in RCA: 128] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 10/30/2017] [Accepted: 11/16/2017] [Indexed: 02/06/2023]
Abstract
The human gut contains a plethora of microbes, providing a platform for metabolic interaction between the host and microbiota. Metabolites produced by the gut microbiota act as a link between gut microbiota and its host. These metabolites act as messengers having the capacity to alter the gut microbiota. Recent advances in the characterization of the gut microbiota and its symbiotic relationship with the host have provided a platform to decode metabolic interactions. The human gut microbiota, a crucial component for dietary metabolism, is shaped by the genetic, epigenetic and dietary factors. The metabolic potential of gut microbiota explains its significance in host health and diseases. The knowledge of interactions between microbiota and host metabolism, as well as modification of microbial ecology, is really beneficial to have effective therapeutic treatments for many diet-related diseases in near future. This review cumulates the information to map the role of human gut microbiota in dietary component metabolism, the role of gut microbes derived metabolites in human health and host-microbe metabolic interactions in health and diseases.
Collapse
Affiliation(s)
- Monika Yadav
- Department of Biochemistry, Maharshi Dayanand University, Rohtak, Haryana, 124001, India
| | - Manoj Kumar Verma
- Department of Biochemistry, Maharshi Dayanand University, Rohtak, Haryana, 124001, India
| | - Nar Singh Chauhan
- Department of Biochemistry, Maharshi Dayanand University, Rohtak, Haryana, 124001, India.
| |
Collapse
|
46
|
Caminer AC, Haberman R, Scher JU. Human microbiome, infections, and rheumatic disease. Clin Rheumatol 2017; 36:2645-2653. [PMID: 29101674 DOI: 10.1007/s10067-017-3875-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 10/05/2017] [Accepted: 10/08/2017] [Indexed: 12/16/2022]
Abstract
Microbes have coevolved with their human hosts for millions of years and are vital to their normal development and homoeostasis. It is now clear that there is direct and indirect cross talk between the microbiome and host immune responses. However, the exact mechanisms for this microbial influence in disease pathogenesis remain elusive and are now a major research focus.
Collapse
Affiliation(s)
- Ana Clara Caminer
- Sanatorio Parque, Universidad Nacional de Rosario, Rosario, Santa Fe, Argentina
| | - Rebecca Haberman
- Psoriatic Arthritis Center, Division of Rheumatology, New York University Langone Health, New York, NY, USA
| | - Jose U Scher
- Psoriatic Arthritis Center, Division of Rheumatology, New York University Langone Health, New York, NY, USA. .,Microbiome Center for Rheumatolgy and Autoimmunity (MiCRA), Division of Rheumatology, New York University School of Medicine and Hospital for Joint Diseases, New York, NY, USA. .,Division of Rheumatology, NYU Hospital for Joint Diseases, Room 1608, 301 East 17th Street, New York, NY, 10003, USA.
| |
Collapse
|
47
|
Jethwa H, Abraham S. The evidence for microbiome manipulation in inflammatory arthritis. Rheumatology (Oxford) 2017; 56:1452-1460. [PMID: 27789760 DOI: 10.1093/rheumatology/kew374] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Indexed: 01/04/2023] Open
Abstract
The human body consists of millions of commensal bacteria (the microbiome), with the intestinal tract being the most prevalent site of colonization. This colonization process begins at birth, and despite numerous factors such as ageing, diet and drug use affecting the microbiome make-up, by adulthood the composition of the gut bacteria is relatively consistent across local populations. The recent advent of new scientific techniques has enabled us to explore how the microbiome affects health and, in particular, has shed light on the involvement of the microbiome in the pathogenesis of inflammatory disease. In this review we highlight the current evidence for microbiome manipulation in inflammatory arthritis in animal and human models and discuss potential therapeutics targeting the microbiome as treatment for these diseases.
Collapse
Affiliation(s)
- Hannah Jethwa
- Rheumatology Department, Wexham Park Hospital, Slough
| | - Sonya Abraham
- NIHR/Wellcome Trust Imperial Clinical Research Facility, Imperial College London, Hammersmith Hospital, London, UK
| |
Collapse
|
48
|
Yan D, Issa N, Afifi L, Jeon C, Chang HW, Liao W. The Role of the Skin and Gut Microbiome in Psoriatic Disease. CURRENT DERMATOLOGY REPORTS 2017; 6:94-103. [PMID: 28804689 DOI: 10.1007/s13671-017-0178-5] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE To understand the changes in the microbiome in psoriatic disease, we conducted a systematic review of studies comparing the skin and gut microbiota in psoriatic individuals and healthy controls. FINDINGS Our review of studies pertaining to the cutaneous microbiome showed a trend towards an increased relative abundance of Streptococcus and a decreased level of Propionibacterium in psoriasis patients compared to controls. In the gut microbiome, the ratio of Firmicutes and Bacteroidetes was perturbed in psoriatic individuals compared to healthy controls. Actinobacteria was also relatively underrepresented in psoriasis patients relative to healthy individuals. SUMMARY Although the field of the psoriatic microbiome is relatively new, these first studies reveal interesting differences in microbiome composition that may be associated with the development of psoriatic comorbidities and serve as novel therapeutic targets.
Collapse
Affiliation(s)
- Di Yan
- Department of Dermatology, University of California-San Francisco, San Francisco, CA, USA
| | - Naiem Issa
- Georgetown University, School of Medicine, Washington D.C., USA
| | - Ladan Afifi
- Department of Dermatology, University of California-San Francisco, San Francisco, CA, USA
| | - Caleb Jeon
- Department of Dermatology, University of California-San Francisco, San Francisco, CA, USA
| | - Hsin Wen Chang
- Department of Dermatology, University of California-San Francisco, San Francisco, CA, USA
| | - Wilson Liao
- Department of Dermatology, University of California-San Francisco, San Francisco, CA, USA
| |
Collapse
|
49
|
Corrêa JD, Calderaro DC, Ferreira GA, Mendonça SMS, Fernandes GR, Xiao E, Teixeira AL, Leys EJ, Graves DT, Silva TA. Subgingival microbiota dysbiosis in systemic lupus erythematosus: association with periodontal status. MICROBIOME 2017; 5:34. [PMID: 28320468 PMCID: PMC5359961 DOI: 10.1186/s40168-017-0252-z] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 03/07/2017] [Indexed: 05/05/2023]
Abstract
BACKGROUND Periodontitis results from the interaction between a subgingival biofilm and host immune response. Changes in biofilm composition are thought to disrupt homeostasis between the host and subgingival bacteria resulting in periodontal damage. Chronic systemic inflammatory disorders have been shown to affect the subgingival microbiota and clinical periodontal status. However, this relationship has not been examined in subjects with systemic lupus erythematosus (SLE). The objective of our study was to investigate the influence of SLE on the subgingival microbiota and its connection with periodontal disease and SLE activity. METHODS We evaluated 52 patients with SLE compared to 52 subjects without SLE (control group). Subjects were classified as without periodontitis and with periodontitis. Oral microbiota composition was assessed by amplifying the V4 region of 16S rRNA gene from subgingival dental plaque DNA extracts. These amplicons were examined by Illumina MiSeq sequencing. RESULTS SLE patients exhibited higher prevalence of periodontitis which occurred at a younger age compared to subjects of the control group. More severe forms of periodontitis were found in SLE subjects that had higher bacterial loads and decreased microbial diversity. Bacterial species frequently detected in periodontal disease were observed in higher proportions in SLE patients, even in periodontal healthy sites such as Fretibacterium, Prevotella nigrescens, and Selenomonas. Changes in the oral microbiota were linked to increased local inflammation, as demonstrated by higher concentrations of IL-6, IL-17, and IL-33 in SLE patients with periodontitis. CONCLUSIONS SLE is associated with differences in the composition of the microbiota, independently of periodontal status.
Collapse
Affiliation(s)
- Jôice Dias Corrêa
- Faculty of Dentistry, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais Brazil
| | | | | | | | - Gabriel R. Fernandes
- René Rachou Research Center, Oswaldo Cruz Foundation, Belo Horizonte, Minas Gerais Brazil
| | - E. Xiao
- Penn Dental School, University of Pennsylvania, Philadelphia, PA USA
| | - Antônio Lúcio Teixeira
- University Hospital, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais Brazil
| | - Eugene J. Leys
- College of Dentistry, The Ohio State University, Columbus, OH USA
| | - Dana T. Graves
- Penn Dental School, University of Pennsylvania, Philadelphia, PA USA
| | - Tarcília Aparecida Silva
- Faculty of Dentistry, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais Brazil
- Departamento de Patologia e Cirurgia Odontológica, Faculdade de Odontologia, Universidade Federal de Minas Gerais, Av. Antônio Carlos 6627, CEP 31.270-901 Belo Horizonte, Minas Gerais Brazil
| |
Collapse
|
50
|
Esmaeili SA, Mahmoudi M, Momtazi AA, Sahebkar A, Doulabi H, Rastin M. Tolerogenic probiotics: potential immunoregulators in Systemic Lupus Erythematosus. J Cell Physiol 2017; 232:1994-2007. [PMID: 27996081 DOI: 10.1002/jcp.25748] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 12/19/2016] [Indexed: 12/15/2022]
Abstract
Probiotics are commensal or nonpathogenic microbes that colonize the gastrointestinal tract and confer beneficial effects on the host through several mechanisms such as competitive exclusion, anti-bacterial effects, and modulation of immune responses. There is growing evidence supporting the immunomodulatory ability of some probiotics. Several experimental and clinical studies have been shown beneficial effect of some probiotic bacteria, particularly Lactobacillus and Bifidobacteria strains, on inflammatory and autoimmune diseases. Systemic lupus erythematosus (SLE) is an autoimmune disease that is mainly characterized by immune intolerance towards self-antigens. Some immunomodulatory probiotics have been found to regulate immune responses via tolerogenic mechanisms. Dendritic and T regulatory (Treg) cells, IL-6, IFN-γ, IL-17, and IL-23 can be considered as the most determinant dysregulated mediators in tolerogenic status. As demonstrated by documented experimental and clinical trials on inflammatory and autoimmune diseases, a number of probiotic bacterial strains can restore tolerance in host through modification of such dysregulated mediators. Since there are limited reports regarding to impact of probiotic supplementation in SLE patients, the preset review was aimed to suggest a number of probiotics bacteria, mainly from Bifidobacteria and Lactobacillus strains that are able to ameliorate immune responses. The aim was followed through literature survey on immunoregulatory probiotics that can restore tolerance and also modulate the important dysregulated pro/anti-inflammatory cytokines contributing to the pathogenesis of SLE.
Collapse
Affiliation(s)
- Seyed-Alireza Esmaeili
- Immunology Research Center, BuAli Research Institute, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Mahmoudi
- Immunology Research Center, BuAli Research Institute, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Abbas Momtazi
- Student Research Committee, Nanotechnology Research Center, Department of Medical Biotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hassan Doulabi
- Immunology Research Center, BuAli Research Institute, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Rastin
- Immunology Research Center, BuAli Research Institute, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|