51
|
Akkermansia muciniphila and Faecalibacterium prausnitzii in Immune-Related Diseases. Microorganisms 2022; 10:microorganisms10122382. [PMID: 36557635 PMCID: PMC9782003 DOI: 10.3390/microorganisms10122382] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/27/2022] [Accepted: 11/28/2022] [Indexed: 12/04/2022] Open
Abstract
Probiotics and synbiotics are used to treat chronic illnesses due to their roles in immune system modulation and anti-inflammatory response. They have been shown to reduce inflammation in a number of immune-related disorders, including systemic lupus erythematosus (SLE), human immunodeficiency virus (HIV), and chronic inflammatory skin conditions such as psoriasis and atopic dermatitis (AD). Akkermansia muciniphila (A. muciniphila) and Faecalibacterium prausnitzii (F. prausnitzii) are two different types of bacteria that play a significant part in this function. It has been established that Akkermansia and Faecalibacterium are abundant in normal populations and have protective benefits on digestive health while also enhancing the immune system, metabolism, and gut barrier of the host. They have the potential to be a therapeutic target in diseases connected to the microbiota, such as immunological disorders and cancer immunotherapy. There has not been a review of the anti-inflammatory effects of Akkermansia and Faecalibacterium, particularly in immunological diseases. In this review, we highlight the most recent scientific findings regarding A. muciniphila and F. prausnitzii as two significant gut microbiota for microbiome alterations and seek to provide cutting-edge insight in terms of microbiome-targeted therapies as promising preventive and therapeutic tools in immune-related diseases and cancer immunotherapy.
Collapse
|
52
|
Widhani A, Djauzi S, Suyatna FD, Dewi BE. Changes in Gut Microbiota and Systemic Inflammation after Synbiotic Supplementation in Patients with Systemic Lupus Erythematosus: A Randomized, Double-Blind, Placebo-Controlled Trial. Cells 2022; 11:3419. [PMID: 36359816 PMCID: PMC9658918 DOI: 10.3390/cells11213419] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 10/22/2022] [Accepted: 10/23/2022] [Indexed: 08/04/2023] Open
Abstract
Gut dysbiosis has a role in the pathogenesis of lupus. Synbiotic supplementation may restore the balance of gut microbiota. This study investigated whether synbiotics could improve gut microbiota and systemic inflammation in lupus patients. This randomized, double-blind, placebo-controlled trial was conducted in adult systemic lupus erythematosus (SLE) patients. Subjects were randomized to receive either synbiotics or a placebo. Fecal microbiota, hs-CRP, IL-6, and IL-17 were measured at baseline and after 60 days. Patients who fulfilled the inclusion criteria were randomized into synbiotic (n = 23) and placebo groups (n = 23). In the synbiotic group, hs-CRP was not significantly increased (1.8 [0.9; 4.85] vs. 2.1 [0.9; 4.25] mg/L; pre vs. post; p = 0.23), whereas in the placebo group hs-CRP was increased significantly (1.75 [0.4; 4.45] vs. 3.75 [0.58; 7.05] mg/L; pre vs. post; p = 0.005). In the synbiotic group, IL-6 decreased significantly (8.76 [6.62; 11.39] vs. 6.59 [4.96; 8.01]; pre vs. post; p = 0.02), while there was no significant change in IL-17 level. In the placebo group, there was no significant change in IL-6 and IL-17. Synbiotic supplementation increased the Firmicutes:Bacteroidetes ratio (0.05 ± 0.60 vs. -0.08 ± 0.63, synbiotic vs. placebo p = 0.48) and butyrate metabolism (p = 0.037) and decreased amino sugar and nucleotide sugar metabolism (p = 0.040). There was improvement in the SLE disease activity index 2K (SLEDAI-2K) score in the synbiotic group (14 [9; 16] vs. 8 [2; 12]; pre vs. post; p < 0.001), while no change in the placebo group (9 [8; 18.25] vs. 9 [5.5; 15]; pre vs. post; p = 0.31). Synbiotic supplementation could reduce systemic inflammation and SLE disease activity and alter the composition and functions of gut microbiota.
Collapse
Affiliation(s)
- Alvina Widhani
- Allergy and Clinical Immunology Division, Department of Internal Medicine, Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia
- Dr. Cipto Mangunkusumo Hospital, Jakarta 10430, Indonesia
- Doctoral Program in Biomedical Science, Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia
| | - Samsuridjal Djauzi
- Allergy and Clinical Immunology Division, Department of Internal Medicine, Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia
- Dr. Cipto Mangunkusumo Hospital, Jakarta 10430, Indonesia
| | | | - Beti Ernawati Dewi
- Department of Microbiology, Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia
| |
Collapse
|
53
|
Zhan Y, Liu Q, Zhang B, Huang X, Lu Q. Recent advances in systemic lupus erythematosus and microbiota: from bench to bedside. Front Med 2022; 16:686-700. [DOI: 10.1007/s11684-022-0957-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 08/18/2022] [Indexed: 11/19/2022]
|
54
|
Wang L, Xu H, Yang H, Zhou J, Zhao L, Zhang F. Glucose metabolism and glycosylation link the gut microbiota to autoimmune diseases. Front Immunol 2022; 13:952398. [PMID: 36203617 PMCID: PMC9530352 DOI: 10.3389/fimmu.2022.952398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 08/19/2022] [Indexed: 11/21/2022] Open
Abstract
Carbohydrates serve as important energy sources and structural substances for human body as well as for gut microbes. As evidenced by the advances in immunometabolism, glucose metabolism and adenosine triphosphate (ATP) generation are deeply involved in immune cell activation, proliferation, and signaling transduction as well as trafficking and effector functions, thus contributing to immune response programming and assisting in host adaption to microenvironment changes. Increased glucose uptake, aberrant expression of glucose transporter 1 (e.g., GLU1), and abnormal glycosylation patterns have been identified in autoimmunity and are suggested as partially responsible for the dysregulated immune response and the modification of gut microbiome composition in the autoimmune pathogenesis. The interaction between gut microbiota and host carbohydrate metabolism is complex and bidirectional. Their impact on host immune homeostasis and the development of autoimmune diseases remains to be elucidated. This review summarized the current knowledge on the crosstalk of glucose metabolism and glycosylation in the host with intestinal microbiota and discussed their possible role in the development and progression of autoimmune diseases. Potential therapeutic strategies targeting glucose metabolism and glycosylation in modulating gut ecosystem and treating autoimmune diseases were discussed as well.
Collapse
Affiliation(s)
- Lu Wang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- Key Laboratory of Rheumatology and Clinical Rheumatology, Ministry of Education, Beijing, China
| | - Haojie Xu
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- Key Laboratory of Rheumatology and Clinical Rheumatology, Ministry of Education, Beijing, China
| | - Huaxia Yang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- Key Laboratory of Rheumatology and Clinical Rheumatology, Ministry of Education, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing, China
| | - Jiaxin Zhou
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- Key Laboratory of Rheumatology and Clinical Rheumatology, Ministry of Education, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing, China
- *Correspondence: Jiaxin Zhou, ; Lidan Zhao,
| | - Lidan Zhao
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- Key Laboratory of Rheumatology and Clinical Rheumatology, Ministry of Education, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing, China
- *Correspondence: Jiaxin Zhou, ; Lidan Zhao,
| | - Fengchun Zhang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- Key Laboratory of Rheumatology and Clinical Rheumatology, Ministry of Education, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing, China
| |
Collapse
|
55
|
Belvoncikova P, Maronek M, Gardlik R. Gut Dysbiosis and Fecal Microbiota Transplantation in Autoimmune Diseases. Int J Mol Sci 2022; 23:10729. [PMID: 36142642 PMCID: PMC9503867 DOI: 10.3390/ijms231810729] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/09/2022] [Accepted: 09/13/2022] [Indexed: 11/16/2022] Open
Abstract
Gut microbiota dysbiosis has recently been reported in a number of clinical states, including neurological, psychiatric, cardiovascular, metabolic and autoimmune disorders. Yet, it is not completely understood how colonizing microorganisms are implicated in their pathophysiology and molecular pathways. There are a number of suggested mechanisms of how gut microbiota dysbiosis triggers or sustains extraintestinal diseases; however, none of these have been widely accepted as part of the disease pathogenesis. Recent studies have proposed that gut microbiota and its metabolites could play a pivotal role in the modulation of immune system responses and the development of autoimmunity in diseases such as rheumatoid arthritis, multiple sclerosis or type 1 diabetes. Fecal microbiota transplantation (FMT) is a valuable tool for uncovering the role of gut microbiota in the pathological processes. This review aims to summarize the current knowledge about gut microbiota dysbiosis and the potential of FMT in studying the pathogeneses and therapies of autoimmune diseases. Herein, we discuss the extraintestinal autoimmune pathologies with at least one published or ongoing FMT study in human or animal models.
Collapse
Affiliation(s)
| | | | - Roman Gardlik
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, 811 08 Bratislava, Slovakia
| |
Collapse
|
56
|
Yaigoub H, Fath N, Tirichen H, Wu C, Li R, Li Y. Bidirectional crosstalk between dysbiotic gut microbiota and systemic lupus erythematosus: What is new in therapeutic approaches? Clin Immunol 2022; 244:109109. [PMID: 36087683 DOI: 10.1016/j.clim.2022.109109] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/25/2022] [Accepted: 08/30/2022] [Indexed: 11/18/2022]
Abstract
Systemic lupus erythematosus is an autoimmune disease characterized by chronic inflammation and multiple organs damage. Its pathogenesis is complex and involves multiple factors including gut microbiota. Accumulating evidence indicates the interaction of microbial communities with the host immune system to maintain a state of homeostasis. Imbalances within the gut microbial composition and function may contribute to the development of many autoimmune diseases including SLE. In this review, we aim to highlight the dysregulation of commensal bacteria and their metabolites in the gastrointestinal tract and the resulting autoimmune responses in lupus and to decrypt the cross-link between the altered gut microbiota and the immune system in the SLE condition. We also provide new insights into targeting gut microbiota as a promising therapeutic approach to treat and manage SLE.
Collapse
Affiliation(s)
- Hasnaa Yaigoub
- Institutes of Biomedical Sciences, Shanxi University, Taiyuan, China
| | - Nada Fath
- Comparative Anatomy Unit, Department of Biological and Pharmacological Veterinary Sciences, Hassan II Agronomy and Veterinary Medicine Institute, Rabat-Instituts, Rabat, Morocco
| | - Hasna Tirichen
- Institutes of Biomedical Sciences, Shanxi University, Taiyuan, China
| | - Changxin Wu
- Institutes of Biomedical Sciences, Shanxi University, Taiyuan, China
| | - Rongshan Li
- Department of Nephrology, The Fifth Hospital (Shanxi Provincial People's Hospital) of Shanxi Medical University, Taiyuan, China; Shanxi Provincial Key Laboratory of Kidney Disease, Taiyuan, China
| | - Yafeng Li
- Department of Nephrology, The Fifth Hospital (Shanxi Provincial People's Hospital) of Shanxi Medical University, Taiyuan, China; Shanxi Provincial Key Laboratory of Kidney Disease, Taiyuan, China; Academy of Microbial Ecology, Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
57
|
Chen Y, Lin J, Xiao L, Zhang X, Zhao L, Wang M, Li L. Gut microbiota in systemic lupus erythematosus: A fuse and a solution. J Autoimmun 2022; 132:102867. [PMID: 35932662 DOI: 10.1016/j.jaut.2022.102867] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 07/05/2022] [Indexed: 12/13/2022]
Abstract
Gut commensals help shape and mold host immune system and deeply influence human health. The disease spectrum of mankind that gut microbiome may associate with is ever-growing, but the mechanisms are still enigmas. Characterized by loss of self-tolerance and sustained self-attack, systemic lupus erythematosus (SLE) is labeled with chronic inflammation, production of autoantibodies and multisystem injury, which so far are mostly incurable. Gut microbiota and their metabolites, now known as important environmental triggers of local/systemic immune responses, have been proposed to be involved in SLE development and progression probably through the following mechanisms: translocation beyond their niches; molecular mimicry to cross-activate immune response targeting self-antigens; epitope spreading to expand autoantibodies spectrum; and bystander activation to promote systemic inflammation. Gut microbiota which varies between individuals may also influence the metabolism and bio-transformation of disease-modifying anti-rheumatic drugs, thus associated with the efficacy and toxicity of these drugs, adding another explanation for heterogenic therapeutic responses. Modulation of gut microbiota via diet, probiotics/prebiotics, antibiotics/phages, fecal microbiota transplantation, or helminth to restore immune tolerance and homeostasis is expected to be a promising neoadjuvant therapy for SLE. We reviewed the advances in this territory and discussed the application prospect of modulating gut microbiota in controlling SLE.
Collapse
Affiliation(s)
- Yanfei Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, China; Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China
| | - Jin Lin
- Department of Rheumatology, The First Affiliated Hospital, College of Medicine, Zhejiang University, China
| | - Lanlan Xiao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, China; Department of Rheumatology, The First Affiliated Hospital, College of Medicine, Zhejiang University, China
| | - Xuan Zhang
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Clinical Immunology Center, Chinese Academy of Medical Sciences & Peking Union Medical College, NO.1 Da Hua Road, Dong Dan, Beijing, 100730, China
| | - Lidan Zhao
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing, 100730, China
| | - Min Wang
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Clinical Immunology Center, Chinese Academy of Medical Sciences & Peking Union Medical College, NO.1 Da Hua Road, Dong Dan, Beijing, 100730, China.
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, China; Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China.
| |
Collapse
|
58
|
Toumi E, Goutorbe B, Plauzolles A, Bonnet M, Mezouar S, Militello M, Mege JL, Chiche L, Halfon P. Gut microbiota in systemic lupus erythematosus patients and lupus mouse model: a cross species comparative analysis for biomarker discovery. Front Immunol 2022; 13:943241. [PMID: 35983031 PMCID: PMC9378784 DOI: 10.3389/fimmu.2022.943241] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 07/04/2022] [Indexed: 11/15/2022] Open
Abstract
An increasing number of studies have provided strong evidence that gut microbiota interact with the immune system and stimulate various mechanisms involved in the pathogenesis of auto-immune diseases such as Systemic Lupus Erythematosus (SLE). Indeed, gut microbiota could be a source of diagnostic and prognostic biomarkers but also hold the promise to discover novel therapeutic strategies. Thus far, specific SLE microbial signatures have not yet been clearly identified with alteration patterns that may vary between human and animal studies. In this study, a comparative analysis of a clinically well-characterized cohort of adult patients with SLE showed reduced biodiversity, a lower Firmicutes/Bacteroidetes (F/B) ratio, and six differentially abundant taxa compared with healthy controls. An unsupervised clustering of patients with SLE patients identified a subgroup of patients with a stronger alteration of their gut microbiota. Interestingly, this clustering was strongly correlated with the disease activity assessed with the Systemic Lupus Erythematosus Disease Activity Index (SLEDAI) score (p = 0.03, odd ratio = 15) and the identification of specific alterations involving the F/B ratio and some different taxa. Then, the gut microbiota of pristane-induced lupus and control mice were analyzed for comparison with our human data. Among the six differentially abundant taxa of the human disease signature, five were common with our murine model. Finally, an exhaustive cross-species comparison between our data and previous human and murine SLE studies revealed a core-set of gut microbiome species that might constitute biomarker panels relevant for future validation studies.
Collapse
Affiliation(s)
- Eya Toumi
- Aix-Marseille Univ, Microbes, Evolution, Phylogénie et infection (MEPHI), Institut de recherche pour le développement (IRD), Assistance Publique-Hopitaux de Marseille (APHM), Marseille, France
- Institut Hospitalo-universaire (IHU)-Méditerranée Infection, Marseille, France
- Laboratoire Alphabio, Clinical Research and R&D Department, Marseille, France
- *Correspondence: Eya Toumi,
| | - Benoit Goutorbe
- Laboratoire Alphabio, Clinical Research and R&D Department, Marseille, France
- Centre de Recherche en Cancérologie de Marseille (CRCM), Aix‐Marseille Univ U105, Inserm U1068, CNRS UMR7258, Institut Paoli‐Calmettes, Marseille, France
- Université Paris-Saclay, Institut national de recherche pour l’agriculture, l’alimentation et l’environnement (INRAE), Mathématiques et Informatique Appliquées du Génome à l’Environnement (MaIAGE), Jouy-en-Josas, France
| | - Anne Plauzolles
- Laboratoire Alphabio, Clinical Research and R&D Department, Marseille, France
| | - Marion Bonnet
- Laboratoire Alphabio, Clinical Research and R&D Department, Marseille, France
| | - Soraya Mezouar
- Aix-Marseille Univ, Microbes, Evolution, Phylogénie et infection (MEPHI), Institut de recherche pour le développement (IRD), Assistance Publique-Hopitaux de Marseille (APHM), Marseille, France
- Institut Hospitalo-universaire (IHU)-Méditerranée Infection, Marseille, France
| | - Muriel Militello
- Aix-Marseille Univ, Microbes, Evolution, Phylogénie et infection (MEPHI), Institut de recherche pour le développement (IRD), Assistance Publique-Hopitaux de Marseille (APHM), Marseille, France
- Institut Hospitalo-universaire (IHU)-Méditerranée Infection, Marseille, France
| | - Jean-Louis Mege
- Aix-Marseille Univ, Microbes, Evolution, Phylogénie et infection (MEPHI), Institut de recherche pour le développement (IRD), Assistance Publique-Hopitaux de Marseille (APHM), Marseille, France
- Institut Hospitalo-universaire (IHU)-Méditerranée Infection, Marseille, France
- Hopital de la Conception, Immunology Department, Marseille, France
| | - Laurent Chiche
- Infectious and Internal Medicine Department, Hôpital Européen Marseille, Marseille, France
| | - Philippe Halfon
- Aix-Marseille Univ, Microbes, Evolution, Phylogénie et infection (MEPHI), Institut de recherche pour le développement (IRD), Assistance Publique-Hopitaux de Marseille (APHM), Marseille, France
- Institut Hospitalo-universaire (IHU)-Méditerranée Infection, Marseille, France
- Laboratoire Alphabio, Clinical Research and R&D Department, Marseille, France
- Infectious and Internal Medicine Department, Hôpital Européen Marseille, Marseille, France
| |
Collapse
|
59
|
Charoensappakit A, Sae-Khow K, Leelahavanichkul A. Gut Barrier Damage and Gut Translocation of Pathogen Molecules in Lupus, an Impact of Innate Immunity (Macrophages and Neutrophils) in Autoimmune Disease. Int J Mol Sci 2022; 23:ijms23158223. [PMID: 35897790 PMCID: PMC9367802 DOI: 10.3390/ijms23158223] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 07/25/2022] [Accepted: 07/25/2022] [Indexed: 02/08/2023] Open
Abstract
The gut barrier is a single cell layer that separates gut micro-organisms from the host, and gut permeability defects result in the translocation of microbial molecules from the gut into the blood. Despite the silent clinical manifestation, gut translocation of microbial molecules can induce systemic inflammation that might be an endogenous exacerbating factor of systemic lupus erythematosus. In contrast, circulatory immune-complex deposition and the effect of medications on the gut, an organ with an extremely large surface area, of patients with active lupus might cause gut translocation of microbial molecules, which worsens lupus severity. Likewise, the imbalance of gut microbiota may initiate lupus and/or interfere with gut integrity which results in microbial translocation and lupus exacerbation. Moreover, immune hyper-responsiveness of innate immune cells (macrophages and neutrophils) is demonstrated in a lupus model from the loss of inhibitory Fc gamma receptor IIb (FcgRIIb), which induces prominent responses through the cross-link between activating-FcgRs and innate immune receptors. The immune hyper-responsiveness can cause cell death, especially apoptosis and neutrophil extracellular traps (NETosis), which possibly exacerbates lupus, partly through the enhanced exposure of the self-antigens. Leaky gut monitoring and treatments (such as probiotics) might be beneficial in lupus. Here, we discuss the current information on leaky gut in lupus.
Collapse
Affiliation(s)
- Awirut Charoensappakit
- Center of Excellence in Translational Research in Inflammation and Immunology (CETRII), Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Kritsanawan Sae-Khow
- Center of Excellence in Translational Research in Inflammation and Immunology (CETRII), Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Asada Leelahavanichkul
- Center of Excellence in Translational Research in Inflammation and Immunology (CETRII), Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
- Nephrology Unit, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
60
|
Wang X, Shu Q, Song L, Liu Q, Qu X, Li M. Gut Microbiota in Systemic Lupus Erythematosus and Correlation With Diet and Clinical Manifestations. Front Med (Lausanne) 2022; 9:915179. [PMID: 35847775 PMCID: PMC9279557 DOI: 10.3389/fmed.2022.915179] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 06/09/2022] [Indexed: 11/25/2022] Open
Abstract
Despite the existing studies relating systemic lupus erythematosus (SLE) to changes in gut microbiota, the latter is affected by external factors such as diet and living environment. Herein, we compared the diversity and composition of gut microbiota in SLE patients and in their healthy family members who share the same household, to link gut microbiota, diet and SLE clinical manifestations. The study cohort included 19 patients with SLE and 19 of their healthy family members. Daily nutrition was assessed using a food frequency questionnaire (FFQ). Microbiota was analyzed using amplicons from the V4 regions of the 16S rRNA gene, to obtain microbiota diversity, taxa relative abundances and network analysis. The gut microbiota in the SLE group had lower alpha diversity and higher heterogeneity than the control group. SLE patients had decreased Acidobacteria, Gemmatimonadetes, Nitrospirae and Planctomycetes at the phylum level, and increased Streptococcus, Veillonella, Clostridium_XI, and Rothia at the genus level. Streptococcus was extremely enriched among patients with lupus nephritis. Lactobacillus, Clostridium_XlVa, Lachnospiracea_incertae_sedis and Parasutterella OTUs were associated with diet and clinical features of SLE. Finally, the gut microbiota of SLE patients remained different from that in healthy controls even after accounting for living conditions and diet.
Collapse
Affiliation(s)
- Xiao Wang
- Department of Rheumatology, Qilu Hospital of Shandong University, Jinan, China
- Shandong Province Clinical Research Center for Immune Diseases and Gout, Jinan, China
| | - Qiang Shu
- Department of Rheumatology, Qilu Hospital of Shandong University, Jinan, China
- Shandong Province Clinical Research Center for Immune Diseases and Gout, Jinan, China
| | - Lijun Song
- Department of Rheumatology, Qilu Hospital of Shandong University, Jinan, China
- Shandong Province Clinical Research Center for Immune Diseases and Gout, Jinan, China
| | - Qi Liu
- Department of Rheumatology, Qilu Hospital of Shandong University, Jinan, China
- Shandong Province Clinical Research Center for Immune Diseases and Gout, Jinan, China
| | - Xiaoxia Qu
- Department of Rheumatology, Qilu Hospital of Shandong University, Jinan, China
- Shandong Province Clinical Research Center for Immune Diseases and Gout, Jinan, China
| | - Ming Li
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, China
- *Correspondence: Ming Li
| |
Collapse
|
61
|
Shahi SK, Yadav M, Ghimire S, Mangalam AK. Role of the gut microbiome in multiple sclerosis: From etiology to therapeutics. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2022; 167:185-215. [PMID: 36427955 DOI: 10.1016/bs.irn.2022.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the CNS that affects around one million people in the United States. Predisposition or protection from this disease is linked with both genetic and environmental factors. In recent years, gut microbiome has emerged as an important environmental factor in the pathobiology of MS. The gut microbiome supports various physiologic functions, including the development and maintenance of the host immune system, the perturbation of which is known as dysbiosis and has been linked with multiple diseases including MS. We and others have shown that people with MS (PwMS) have gut dysbiosis that is characterized by specific gut bacteria being enriched or depleted. Consequently, there is an emphasis on determining the mechanism(s) through which gut bacteria and/or their metabolites alter the course of MS through their ability to provide protection, predispose individuals, or promote disease progression. Improving our understanding of these mechanisms will allow us to harness the enormous potential of the gut microbiome as a diagnostic and/or therapeutic agent. In this chapter, we will discuss current advances in microbiome research in the context of MS, including a review of specific bacteria that are currently linked with this disease, potential mechanisms of disease pathogenesis, and the utility of microbiome-based therapy for PwMS.
Collapse
Affiliation(s)
- Shailesh K Shahi
- Department of Pathology, University of Iowa, Iowa City, IA, United States; Iowa City VA Health System, Iowa City, IA, United States
| | - Meeta Yadav
- Department of Pathology, University of Iowa, Iowa City, IA, United States; Iowa City VA Health System, Iowa City, IA, United States
| | - Sudeep Ghimire
- Department of Pathology, University of Iowa, Iowa City, IA, United States; Iowa City VA Health System, Iowa City, IA, United States
| | - Ashutosh K Mangalam
- Department of Pathology, University of Iowa, Iowa City, IA, United States; Iowa City VA Health System, Iowa City, IA, United States.
| |
Collapse
|
62
|
Abstract
Systemic Lupus Erythematosus is a complex autoimmune disease and its etiology remains unknown. Increased gut permeability has been reported in lupus patients, yet whether it promotes or results from lupus progression is unclear. Recent studies indicate that an impaired intestinal barrier allows the translocation of bacteria and bacterial components into systemic organs, increasing immune cell activation and autoantibody generation. Indeed, induced gut leakage in a mouse model of lupus enhanced disease characteristics, including the production of anti-dsDNA antibody, serum IL-6 as well as cell apoptosis. Gut microbiota dysbiosis has been suggested to be one of the factors that decreases gut barrier integrity by outgrowing harmful bacteria and their products, or by perturbation of gut immune homeostasis, which in turn affects gut barrier integrity. The restoration of microbial balance eliminates gut leakage in mice, further confirming the role of microbiota in maintaining gut barrier integrity. In this review, we discuss recent advances on the association between microbiota dysbiosis and leaky gut, as well as their influences on the progression of lupus. The modifications on host microbiota and gut integrity may offer insights into the development of new lupus treatment.
Collapse
Affiliation(s)
| | - Laurence Morel
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL, United States
| |
Collapse
|
63
|
Wang W, Fan Y, Wang X. Lactobacillus: Friend or Foe for Systemic Lupus Erythematosus? Front Immunol 2022; 13:883747. [PMID: 35677055 PMCID: PMC9168270 DOI: 10.3389/fimmu.2022.883747] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 04/26/2022] [Indexed: 12/03/2022] Open
Abstract
The cause of Systemic Lupus Erythematosus (SLE) remains largely unknown, despite the fact that it is well understood that a complex interaction between genes and environment is required for disease development. Microbiota serve as activators and are essential to immune homeostasis. Lactobacillus is thought to be an environmental agent affecting the development of SLE. However, beneficial therapeutic and anti-inflammatory effects of Lactobacillus on SLE were also explored. The discovery of Lactobacillus involvement in SLE will shed light on how SLE develops, as well as finding microbiota-targeted biomarkers and novel therapies. In this review, we attempt to describe the two sides of Lactobacillus in the occurrence, development, treatment and prognosis of SLE. We also discuss the effect of different strains Lactobacillus on immune cells, murine lupus, and patients. Finally, we try to illustrate the potential immunological mechanisms of Lactobacillus on SLE and provide evidence for further microbiota-targeted therapies.
Collapse
Affiliation(s)
- Weijie Wang
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Yongsheng Fan
- School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xinchang Wang
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
64
|
La Barbera L, Macaluso F, Fasano S, Grasso G, Ciccia F, Guggino G. Microbiome Changes in Connective Tissue Diseases and Vasculitis: Focus on Metabolism and Inflammation. Int J Mol Sci 2022; 23:ijms23126532. [PMID: 35742974 PMCID: PMC9224234 DOI: 10.3390/ijms23126532] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/31/2022] [Accepted: 06/06/2022] [Indexed: 02/04/2023] Open
Abstract
The microbial community acts as an active player in maintaining homeostasis and immune functions through a continuous and changeable cross-talk with the host immune system. Emerging evidence suggests that altered microbial composition, known as dysbiosis, might perturb the delicate balance between the microbiota and the immune system, triggering inflammation and potentially contributing to the pathogenesis and development of chronic inflammatory diseases. This review will summarize the current evidence about the microbiome-immunity cross-talk, especially focusing on the microbiota alterations described in patients with rheumatic diseases and on the recent findings concerning the interaction between microbiota, metabolic function, and the immune system.
Collapse
Affiliation(s)
- Lidia La Barbera
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Rheumatology Section, University of Palermo, Piazza delle Cliniche 2, 90110 Palermo, Italy; (L.L.B.); (G.G.)
| | - Federica Macaluso
- Rheumatology Unit, Department of Internal Medicine, University of Modena and Reggio Emilia, AUSL-IRCCS, Via Giovanni Amendola, 2, 42122 Reggio Emilia, Italy;
- Division of Rheumatology, Department of Precision Medicine, University of Campania Luigi Vanvitelli, S. Andrea delle Dame, Via L. De Crecchio 7, 80138 Naples, Italy; (S.F.); (F.C.)
| | - Serena Fasano
- Division of Rheumatology, Department of Precision Medicine, University of Campania Luigi Vanvitelli, S. Andrea delle Dame, Via L. De Crecchio 7, 80138 Naples, Italy; (S.F.); (F.C.)
| | - Giulia Grasso
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Rheumatology Section, University of Palermo, Piazza delle Cliniche 2, 90110 Palermo, Italy; (L.L.B.); (G.G.)
| | - Francesco Ciccia
- Division of Rheumatology, Department of Precision Medicine, University of Campania Luigi Vanvitelli, S. Andrea delle Dame, Via L. De Crecchio 7, 80138 Naples, Italy; (S.F.); (F.C.)
| | - Giuliana Guggino
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Rheumatology Section, University of Palermo, Piazza delle Cliniche 2, 90110 Palermo, Italy; (L.L.B.); (G.G.)
- Correspondence: ; Tel.: +39-091-655-2148
| |
Collapse
|
65
|
Shaheen WA, Quraishi MN, Iqbal TH. Gut microbiome and autoimmune disorders. Clin Exp Immunol 2022; 209:161-174. [PMID: 35652460 DOI: 10.1093/cei/uxac057] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 04/29/2022] [Accepted: 05/30/2022] [Indexed: 12/13/2022] Open
Abstract
Autoimmune diseases have long been known to share a common pathogenesis involving a dysregulated immune system with failure to recognize self from non-self antigens. This immune dysregulation is now increasingly understood to be induced by environmental triggers in genetically predisposed individuals. Although several external environmental triggers have been defined in different autoimmune diseases, much attention is being paid to the role of the internal micro-environment occupied by the microbiome which was once termed "the forgotten organ". In this regard, the gut microbiome, serving as an intermediary between some of those external environmental effectors and the immune system helps programming of the immune system to be tolerant to innocent external and self antigens. However, in the presence of perturbed gut microbiota (dysbiosis), the immune system could be erroneously directed in favor of pro-inflammatory pathways to instigate different autoimmune processes. An accumulating body of evidence, including both experimental and human studies (observational and interventional) points to a role of gut microbiome in different autoimmune diseases. Such evidence could provide a rationale for gut microbiome manipulation with therapeutic and even preventative intents in patients with established or predisposed to autoimmune diseases respectively. Perturbations of the gut microbiome have been delineated in some immune mediated diseases, IBD in particular. However, such patterns of disturbance (microbiome signatures) and related pathogenetic roles of the gut microbiome are context dependent and cannot be generalized in the same exact way to other autoimmune disorders and the contribution of gut microbiome to different disease phenotypes has to be precisely defined. In this review, we revise the evidence for a role of gut microbiome in various autoimmune diseases and possible mechanisms mediating such a role.
Collapse
Affiliation(s)
- Walaa Abdelaty Shaheen
- University of Birmingham Microbiome Treatment Center, Birmingham, UK.,Institute of Cancer and Genomic Sciences, University of Birmingham, UK.,Gastroenterology Department, Menoufia University, Egypt
| | - Mohammed Nabil Quraishi
- University of Birmingham Microbiome Treatment Center, Birmingham, UK.,Institute of Cancer and Genomic Sciences, University of Birmingham, UK.,University Hospitals of Birmingham NHS Foundation Trust, Birmingham, UK
| | - Tariq H Iqbal
- University of Birmingham Microbiome Treatment Center, Birmingham, UK.,Institute of Microbiology and Infection, University of Birmingham, UK.,University Hospitals of Birmingham NHS Foundation Trust, Birmingham, UK
| |
Collapse
|
66
|
James WA, Ogunrinde E, Wan Z, Kamen DL, Oates J, Gilkeson GS, Jiang W. A Distinct Plasma Microbiome But Not Gut Microbiome in Patients With Systemic Lupus Erythematosus Compared to Healthy Individuals. J Rheumatol 2022; 49:592-597. [PMID: 35169056 PMCID: PMC9364828 DOI: 10.3899/jrheum.210952] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/25/2022] [Indexed: 06/14/2023]
Abstract
OBJECTIVE Blood microbiome has been analyzed in cancer patients using machine learning. We aimed to study whether the plasma microbiome represents the microbial community in the gut among patients with systemic lupus erythematosus (SLE) and healthy controls (HCs). METHODS Paired plasma and stool samples from female patients with SLE and female HCs were assessed for microbiome composition by microbial 16S ribosomal RNA sequencing. RESULTS Decreased microbial alpha diversity in stool compared to plasma and distinct plasma and gut beta diversity were found in both HCs and patients with SLE. No difference in gut microbial diversity was found; however, plasma alpha diversity was decreased in patients with SLE compared to HCs. The predominant bacteria differed between plasma and stool in both groups. Although the predominant plasma and stool genus bacteria were similar in patients with SLE and HCs, some were clearly different. CONCLUSION Compared to the gut, the plasma microbiome contained distinct community and greater heterogeneity, indicating that the predominant circulating microbiome may originate from sites (eg, oral or skin) other than the gastrointestinal tract. The decreased plasma but not gut alpha diversity in patients with SLE compared to HCs implies an altered plasma microbiome in SLE, which may be important for systemic immune perturbations and SLE disease pathogenesis.
Collapse
Affiliation(s)
- Warren A James
- W.A. James, MD, College of Medicine, Medical University of South Carolina
| | - Elizabeth Ogunrinde
- E. Ogunrinde, PhD, Z. Wan, MS, Department of Microbiology and Immunology, Medical University of South Carolina
| | - Zhuang Wan
- E. Ogunrinde, PhD, Z. Wan, MS, Department of Microbiology and Immunology, Medical University of South Carolina
| | - Diane L Kamen
- D.L. Kamen, MD, J. Oates, Director and Endowed Chair, MD, Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, and Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina
| | - Jim Oates
- D.L. Kamen, MD, J. Oates, Director and Endowed Chair, MD, Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, and Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina
| | - Gary S Gilkeson
- G.S. Gilkeson, MD, Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, and Staff Physician, Ralph H. Johnson VA Medical Center;
| | - Wei Jiang
- W. Jiang, MD, Department of Microbiology and Immunology, and Division of Infectious Diseases, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA.
| |
Collapse
|
67
|
Brown J, Abboud G, Ma L, Choi SC, Kanda N, Zeumer-Spataro L, Lee J, Peng W, Cagmat J, Faludi T, Mohamadzadeh M, Garrett T, Mandik-Nayak L, Chervonsky A, Perl A, Morel L. Microbiota-mediated skewing of tryptophan catabolism modulates CD4+ T cells in lupus-prone mice. iScience 2022; 25:104241. [PMID: 35494242 PMCID: PMC9051618 DOI: 10.1016/j.isci.2022.104241] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 02/14/2022] [Accepted: 04/07/2022] [Indexed: 12/21/2022] Open
Abstract
A skewed tryptophan metabolism has been reported in patients with lupus. Here, we investigated the mechanisms by which it occurs in lupus-susceptible mice, and how tryptophan metabolites exacerbate T cell activation. Metabolomic analyses demonstrated that tryptophan is differentially catabolized in lupus mice compared to controls and that the microbiota played a role in this skewing. There was no evidence for differential expression of tryptophan catabolic enzymes in lupus mice, further supporting a major contribution of the microbiota to skewing. However, isolated lupus T cells processed tryptophan differently, suggesting a contribution of T cell intrinsic factors. Functionally, tryptophan and its microbial product tryptamine increased T cell metabolism and mTOR activation, while kynurenine promoted interferon gamma production, all of which have been associated with lupus. These results showed that a combination of microbial and T cell intrinsic factors promotes the production of tryptophan metabolites that enhance inflammatory phenotypes in lupus T cells. Intestinal dysbiosis skews tryptophan catabolism in lupus-prone mice Murine lupus CD4+ T cells have an intrinsically different processing of tryptophan Tryptophan and tryptamine increase mTOR activation and metabolism in CD4+ T cells Kynurenine promotes IFNγ production in CD4+ T cells from lupus-prone mice
Collapse
Affiliation(s)
- Josephine Brown
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Georges Abboud
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Longhuan Ma
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Seung-Chul Choi
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Nathalie Kanda
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Leilani Zeumer-Spataro
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Jean Lee
- Committee on Immunology, The University of Chicago, Chicago, IL 60637, USA
| | - Weidan Peng
- Lankenau Institute for Medical Research, Wynnewood, PA, USA
| | - Joy Cagmat
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Tamas Faludi
- Departments of Medicine, Microbiology and Immunology, Biochemistry and Molecular Biology, State University of New York, Upstate Medical University, College of Medicine, Syracuse, NY 13210, USA
| | - Mansour Mohamadzadeh
- Department of Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Timothy Garrett
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610, USA
| | | | | | - Andras Perl
- Departments of Medicine, Microbiology and Immunology, Biochemistry and Molecular Biology, State University of New York, Upstate Medical University, College of Medicine, Syracuse, NY 13210, USA
| | - Laurence Morel
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610, USA
- Corresponding author
| |
Collapse
|
68
|
Li XB, Chu XJ, Cao NW, Wang H, Fang XY, Fan YG, Li BZ, Ye DQ. Proton pump inhibitors induce changes in the gut microbiome composition of systemic lupus erythematosus patients. BMC Microbiol 2022; 22:117. [PMID: 35477382 PMCID: PMC9043501 DOI: 10.1186/s12866-022-02533-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 04/20/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Currently, few studies focus on the association between gut microbiota and systemic lupus erythematosus (SLE), and much less studies consider the effect of drug usage. Proton pump inhibitors (PPIs) are commonly used to treat drug-related gastrointestinal damage in SLE patients. Therefore, the purpose of this study is to examine the gut microbiota of SLE patients using PPIs. METHODS Fecal samples from 20 SLE patients with PPIs (P-SLE), 20 SLE patients without PPIs (NP-SLE) and 17 healthy controls (HCs) were obtained. The structure of the bacterial community in the fecal samples was analyzed by 16S rRNA gene sequencing. Redundancy analysis (RDA) was performed to observe the relationship between clinical variables and microbiome composition in P-SLE and NP-SLE patients. Based on the Kyoto Encyclopedia of Genes and Genomes (KEGG) database, functional capabilities of microbiota were estimated. Network analysis was performed to analyze the association of metabolic pathway alterations with altered gut microbiota in P-SLE and NP-SLE patients. RESULTS P-SLE patients exhibited increased alpha-diversity and an altered composition of the gut microbiota compared with NP-SLE patients. The alpha-diversity of NP-SLE patients was significantly lower than HCs but also of P-SLE patients, whose alpha-diversity had become similar to HCs. Compared with NP-SLE patients, the relative abundances of Lactobacillus, Roseburia, Oxalobacter, and Desulfovibrio were increased, while those of Veillonella, Escherichia, Morganella, Pseudomonas and Stenotrophomonas were decreased in P-SLE patients. RDA indicated that PPI use was the only significant exploratory variable for the microbiome composition when comparing SLE patients. KEGG analysis showed that 16 metabolic pathways were significantly different between NP-SLE and P-SLE patients. These metabolic pathways were mainly associated with changes in Escherichia, Roseburia, Stenotrophomonas, Morganella and Alipipes as determined by the network analysis. CONCLUSIONS PPI use is associated with an improved microbiome composition of SLE patients as it 1) increases alpha-diversity levels back to normal, 2) increases the abundance of various (beneficial) commensals, and 3) decreases the abundance of certain opportunistic pathogenic genera such as Escherichia. Validation studies with higher patient numbers are however recommended to explore these patterns in more detail.
Collapse
Affiliation(s)
- Xian-Bao Li
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Xiu-Jie Chu
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Nv-Wei Cao
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Hua Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Xin-Yu Fang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Yin-Guang Fan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Bao-Zhu Li
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China. .,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China.
| | - Dong-Qing Ye
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China. .,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China.
| |
Collapse
|
69
|
Xiang S, Qu Y, Qian S, Wang R, Wang Y, Jin Y, Li J, Ding X. Association between systemic lupus erythematosus and disruption of gut microbiota: a meta-analysis. Lupus Sci Med 2022; 9:9/1/e000599. [PMID: 35346981 PMCID: PMC8961174 DOI: 10.1136/lupus-2021-000599] [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: 10/09/2021] [Accepted: 03/06/2022] [Indexed: 12/13/2022]
Abstract
Objective Recent studies reported that SLE is characterised by altered interactions between the microbiome and immune system. We performed a meta-analysis of publications on this topic. Methods Case–control studies that compared patients with SLE and healthy controls (HCs) and determined the diversity of the gut microbiota and the abundance of different microbes were examined. Stata/MP V.16 was used for the meta-analysis. A Bonferroni correction for multiple tests was used to reduce the likelihood of false-positive results. Results We included 11 case–control studies that examined 373 patients with SLE and 1288 HCs. These studies were performed in five countries and nine cities. Compared with HCs, patients with SLE had gut microbiota with lower Shannon-Wiener diversity index (weighted mean difference=−0.22, 95% CI −0.32 to –0.13, p<0.001) and lower Chao1 richness (standardised mean difference (SMD)=−0.62, 95% CI −1.04 to –0.21, p=0.003). Patients with SLE had lower abundance of Ruminococcaceae (SMD = −0.49, 95% CI −0.84 to −0.15, p=0.005), but greater abundance of Enterobacteriaceae (SMD=0.45, 95% CI 0.01 to 0.89, p=0.045) and Enterococcaceae (SMD=0.53, 95% CI 0.05 to 1.01, p=0.03). However, only the results for Ruminococcaceae passed the Bonferroni correction (p=0.0071). The two groups had no significant differences in Lachnospiraceae and Bacteroides (both p>0.05). Patients with SLE who used high doses of glucocorticoids had altered gut microbiota based on the Chao1 species diversity estimator, and hydroxychloroquine use appeared to reduce the abundance of Enterobacteriaceae. Conclusions Patients with SLE have imbalanced gut microbiota, with a decrease in beneficial bacteria and an increase in harmful bacteria. Drugs used to treat SLE may also alter the gut microbiota of these patients.
Collapse
Affiliation(s)
- Shate Xiang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yiqian Qu
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Suhai Qian
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Rongyun Wang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yao Wang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yibo Jin
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jie Li
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xinghong Ding
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
70
|
Valiente GR, Munir A, Hart ML, Blough P, Wada TT, Dalan EE, Willis WL, Wu LC, Freud AG, Jarjour WN. Gut dysbiosis is associated with acceleration of lupus nephritis. Sci Rep 2022; 12:152. [PMID: 34996983 PMCID: PMC8742035 DOI: 10.1038/s41598-021-03886-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 12/01/2021] [Indexed: 12/20/2022] Open
Abstract
The gut microbiota (GM) exerts a strong influence over the host immune system and dysbiosis of this microbial community can affect the clinical phenotype in chronic inflammatory conditions. To explore the role of the GM in lupus nephritis, we colonized NZM2410 mice with Segmented Filamentous Bacteria (SFB). Gut colonization with SFB was associated with worsening glomerulonephritis, glomerular and tubular immune complex deposition and interstitial inflammation compared to NZM2410 mice free of SFB. With SFB colonization mice experienced an increase in small intestinal lamina propria Th17 cells and group 3 innate lymphoid cells (ILC3s). However, although serum IL-17A expression was elevated in these mice, Th17 cells and ILC3s were not detected in the inflammatory infiltrate in the kidney. In contrast, serum and kidney tissue expression of the macrophage chemoattractants MCP-1 and CXCL1 were significantly elevated in SFB colonized mice. Furthermore, kidney infiltrating F4/80+CD206+M2-like macrophages were significantly increased in these mice. Evidence of increased gut permeability or "leakiness" was also detected in SFB colonized mice. Finally, the intestinal microbiome of SFB colonized mice at 15 and 30 weeks of age exhibited dysbiosis when compared to uncolonized mice at the same time points. Both microbial relative abundance as well as biodiversity of colonized mice was found to be altered. Collectively, SFB gut colonization in the NZM2410 mouse exacerbates kidney disease, promotes kidney M2-like macrophage infiltration and overall intestinal microbiota dysbiosis.
Collapse
Affiliation(s)
- Giancarlo R Valiente
- Medical Scientist Training Program, The Ohio State University, Columbus, OH, USA
| | - Armin Munir
- Department of Rheumatology and Immunology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | | | - Perry Blough
- Department of Rheumatology and Immunology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Takuma T Wada
- Saitama Medical University, Moroyama, Saitama, Japan
| | - Emma E Dalan
- Department of Rheumatology and Immunology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - William L Willis
- Department of Rheumatology and Immunology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Lai-Chu Wu
- Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH, USA
| | - Aharon G Freud
- The Department of Pathology and the James Cancer Hospital and Solove Research Institute, The Ohio State University, Columbus, OH, USA
| | - Wael N Jarjour
- Department of Rheumatology and Immunology, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
71
|
Abdelhamid L, Luo XM. Diet and Hygiene in Modulating Autoimmunity During the Pandemic Era. Front Immunol 2022; 12:749774. [PMID: 35069526 PMCID: PMC8766844 DOI: 10.3389/fimmu.2021.749774] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 12/13/2021] [Indexed: 12/11/2022] Open
Abstract
The immune system is an efficiently toned machinery that discriminates between friends and foes for achieving both host defense and homeostasis. Deviation of immune recognition from foreign to self and/or long-lasting inflammatory responses results in the breakdown of tolerance. Meanwhile, educating the immune system and developing immunological memory are crucial for mounting defensive immune responses while protecting against autoimmunity. Still to elucidate is how diverse environmental factors could shape autoimmunity. The emergence of a world pandemic such as SARS-CoV-2 (COVID-19) not only threatens the more vulnerable individuals including those with autoimmune conditions but also promotes an unprecedented shift in people's dietary approaches while urging for extraordinary hygiene measures that likely contribute to the development or exacerbation of autoimmunity. Thus, there is an urgent need to understand how environmental factors modulate systemic autoimmunity to better mitigate the incidence and or severity of COVID-19 among the more vulnerable populations. Here, we discuss the effects of diet (macronutrients and micronutrients) and hygiene (the use of disinfectants) on autoimmunity with a focus on systemic lupus erythematosus.
Collapse
Affiliation(s)
- Leila Abdelhamid
- Department of Biomedical Sciences and Pathobiology, College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
- Department of Microbiology, College of Veterinary Medicine, Alexandria University, Alexandria, Egypt
| | - Xin M. Luo
- Department of Biomedical Sciences and Pathobiology, College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| |
Collapse
|
72
|
Tsigalou C, Konstantinidis T, Aloizou AM, Bezirtzoglou E, Tsakris A. Future Therapeutic Prospects in Dealing with Autoimmune Diseases: Treatment Based on the Microbiome Model. ROLE OF MICROORGANISMS IN PATHOGENESIS AND MANAGEMENT OF AUTOIMMUNE DISEASES 2022:489-520. [DOI: 10.1007/978-981-19-4800-8_25] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
73
|
Watane A, Cavuoto KM, Rojas M, Dermer H, Day JO, Banerjee S, Galor A. Fecal Microbial Transplant in Individuals With Immune-Mediated Dry Eye. Am J Ophthalmol 2022; 233:90-100. [PMID: 34214453 PMCID: PMC8678170 DOI: 10.1016/j.ajo.2021.06.022] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 06/01/2021] [Accepted: 06/23/2021] [Indexed: 01/03/2023]
Abstract
PURPOSE To evaluate the safety of the Fecal Microbial Transplant for Sjogren Syndrome (FMT) trial in individuals with immune-mediated dry eye (DE). DESIGN Open-label, nonrandomized clinical trial. METHODS The study population included 10 individuals with DE symptoms and signs meeting criteria for Sjögren or positive early Sjögren markers. Procedures were 2 FMTs from a single healthy donor delivered via enema, 1 week apart. The primary outcome measure was safety. In addition, gut microbiome profiles, DE metrics, and T-cell profiles in blood were examined at baseline before FMT, and at 1 week, 1 month, and 3 months after FMT. RESULTS The mean age of the population was 60.4 years; 30% were male; 50% were white; and 50% were Hispanic. At baseline, all subjects had significantly different gut microbiome profiles from the donor, including higher mean diversity indices. Subjects had a decreased abundance of genera Faecalibacterium, Prevotella, and Ruminococcus and an increased abundance of genera Alistipes, Streptococcus, and Blautia compared to the donor. Effector and regulatory T-cell profiles were positively correlated with each other and with DE symptom severity (T helper 1 cells [Th1]; r = .76; P = .01; Th17: r = 0.83; P = .003; CD25: r = 0.66; P = .04; FoxP3: r = 0.68; P = .03). No adverse events were noted with FMT. After FMT, gut microbiome profiles in 8 subjects moved closer to the donor's profile. As a group, gut microbiome profiles at all follow-up time points were more similar to the original recipients' than the donor's microbiome; however, certain phyla, classes, and genera operational taxonomic unit (OTU) numbers remained closer to the donor vs recipients' baseline profiles out to 3 months. Five individuals subjectively reported improved dry eye symptoms 3 months after FMT. CONCLUSIONS FMT was safely performed in individuals with immune-mediated DE, with certain bacterial profiles resembling the donor out to 3 months after FMT. One-half the subjects reported improved DE symptoms. The most effective FMT administration method has yet to be determined.
Collapse
Affiliation(s)
- Arjun Watane
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL
| | - Kara M. Cavuoto
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL
| | - Mario Rojas
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL
| | - Harrison Dermer
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL
| | - Joanne O Day
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL
| | - Santanu Banerjee
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL
| | - Anat Galor
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL,Miami Veterans Administration Medical Center, 1201 NW 16th St, Miami, FL 33125
| |
Collapse
|
74
|
Pan Q, Guo F, Huang Y, Li A, Chen S, Chen J, Liu HF, Pan Q. Gut Microbiota Dysbiosis in Systemic Lupus Erythematosus: Novel Insights into Mechanisms and Promising Therapeutic Strategies. Front Immunol 2021; 12:799788. [PMID: 34925385 PMCID: PMC8677698 DOI: 10.3389/fimmu.2021.799788] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 11/18/2021] [Indexed: 12/12/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease that was traditionally thought to be closely related to genetic and environmental risk factors. Although treatment options for SLE with hormones, immunosuppressants, and biologic drugs are now available, the rates of clinical response and functional remission of these drugs are still not satisfactory. Currently, emerging evidence suggests that gut microbiota dysbiosis may play crucial roles in the occurrence and development of SLE, and manipulation of targeting the gut microbiota holds great promises for the successful treatment of SLE. The possible mechanisms of gut microbiota dysbiosis in SLE have not yet been well identified to date, although they may include molecular mimicry, impaired intestinal barrier function and leaky gut, bacterial biofilms, intestinal specific pathogen infection, gender bias, intestinal epithelial cells autophagy, and extracellular vesicles and microRNAs. Potential therapies for modulating gut microbiota in SLE include oral antibiotic therapy, fecal microbiota transplantation, glucocorticoid therapy, regulation of intestinal epithelial cells autophagy, extracellular vesicle-derived miRNA therapy, mesenchymal stem cell therapy, and vaccination. This review summarizes novel insights into the mechanisms of microbiota dysbiosis in SLE and promising therapeutic strategies, which may help improve our understanding of the pathogenesis of SLE and provide novel therapies for SLE.
Collapse
Affiliation(s)
- Quanren Pan
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Fengbiao Guo
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Yanyan Huang
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Aifen Li
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Shuxian Chen
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Jiaxuan Chen
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Hua-Feng Liu
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Qingjun Pan
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
75
|
Guo X, Yang X, Li Q, Shen X, Zhong H, Yang Y. The Microbiota in Systemic Lupus Erythematosus: An Update on the Potential Function of Probiotics. Front Pharmacol 2021; 12:759095. [PMID: 34887760 PMCID: PMC8650621 DOI: 10.3389/fphar.2021.759095] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Accepted: 11/01/2021] [Indexed: 12/18/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a kind of chronic diffuse connective tissue illness characterized by multisystem and multiorgan involvement, repeated recurrence and remission, and the presence of a large pool of autoantibodies in the body. Although the exact cause of SLE is not thoroughly revealed, accumulating evidence has manifested that intake of probiotics alters the composition of the gut microbiome, regulating the immunomodulatory and inflammatory response, which may be linked to the disease pathogenesis. Particularly, documented experiments demonstrated that SLE patients have remarkable changes in gut microbiota compared to healthy controls, indicating that the alteration of microbiota may be implicated in different phases of SLE. In this review, the alteration of microbiota in the development of SLE is summarized, and the mechanism of intestinal microbiota on the progression of immune and inflammatory responses in SLE is also discussed. Due to limited reports on the effects of probiotics supplementation in SLE patients, we emphasize advancements made in the last few years on the function and mechanisms of probiotics in the development of SLE animal models. Besides, we follow through literature to survey whether probiotics supplements can be an adjuvant therapy for comprehensive treatment of SLE. Research has indicated that intake of probiotics alters the composition of the gut microbiome, contributing to prevent the progression of SLE. Adjustment of the gut microbiome through probiotics supplementation seems to alleviate SLE symptoms and their cardiovascular and renal complications in animal models, marking this treatment as a potentially novel approach.
Collapse
Affiliation(s)
- Xirui Guo
- Department of Pharmacy, Chengdu Second People's Hospital, Chengdu, China
| | - Xuerong Yang
- Department of Pharmacy, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Qi Li
- Department of Pharmacy, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiaoyan Shen
- Department of Pharmacy, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Huiyun Zhong
- Department of Pharmacy, Sichuan Vocational College of Health and Rehabilitation, Zigong, China.,Department of Pharmacy, The First People's Hospital of Zigong, Zigong, China
| | - Yong Yang
- Department of Pharmacy, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
76
|
Ma Y, Guo R, Sun Y, Li X, He L, Li Z, Silverman GJ, Chen G, Gao F, Yuan J, Wei Q, Li M, Lu L, Niu H. Lupus gut microbiota transplants cause autoimmunity and inflammation. Clin Immunol 2021; 233:108892. [PMID: 34813937 DOI: 10.1016/j.clim.2021.108892] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 11/17/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND The etiology of systemic lupus erythematosus (SLE) is multifactorial. Recently, growing evidence suggests that the microbiota plays a role in SLE, yet whether gut microbiota participates in the development of SLE remains largely unknown. To investigate this issue, we carried out 16 s rDNA sequencing analyses in a cohort of 18 female un-treated active SLE patients and 7 female healthy controls, and performed fecal microbiota transplantation from patients and healthy controls to germ-free (GF) mice. RESULTS Compared to the healthy controls, we found no significant different microbial diversity but some significantly different species in SLE patients including Turicibacter genus and other 5 species. Fecal transfer from SLE patients to GF mice caused GF mice to develop a series of lupus-like phenotypic features, including increased serum autoimmune antibodies, imbalanced cytokines, altered distribution of immune cells in mucosal and peripheral immune response, and upregulated expression of genes related to SLE in recipient mice that received SLE fecal microbiota transplantation (FMT). Moreover, the metabolism of histidine was significantly altered in GF mice treated with SLE patient feces, as compared to those which received healthy fecal transplants. CONCLUSIONS Overall, our results describe a causal role of aberrant gut microbiota in contributing to the pathogenesis of SLE. The interplay of gut microbial and histidine metabolism may be one of the mechanisms intertwined with autoimmune activation in SLE.
Collapse
Affiliation(s)
- Yiyangzi Ma
- School of Medicine and Institute of Laboratory Animal Sciences, Jinan University; Guangzhou Key Laboratory of Germ-free Animals and Microbiota Application, Guangzhou 510632, China; Department of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200001, China
| | - Ruru Guo
- Department of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200001, China
| | - Yiduo Sun
- Department of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences & Peking Union Medical College; Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing 100730, China; Department of Rheumatology,The First Affiliated Hospital, Zhejiang University School of Medicine (FAHZU), Hangzhou 310003, China
| | - Xin Li
- School of Medicine and Institute of Laboratory Animal Sciences, Jinan University; Guangzhou Key Laboratory of Germ-free Animals and Microbiota Application, Guangzhou 510632, China
| | - Lun He
- School of Medicine and Institute of Laboratory Animal Sciences, Jinan University; Guangzhou Key Laboratory of Germ-free Animals and Microbiota Application, Guangzhou 510632, China
| | - Zhao Li
- Department of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences & Peking Union Medical College; Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing 100730, China; Beijing Hospital, National Center of Gerontology, Beijing 100730, China
| | - Gregg J Silverman
- Division of Rheumatology, New York University School of Medicine, New York, NY 10016, USA
| | - Guobing Chen
- School of Medicine and Institute of Laboratory Animal Sciences, Jinan University; Guangzhou Key Laboratory of Germ-free Animals and Microbiota Application, Guangzhou 510632, China
| | - Feng Gao
- School of Medicine and Institute of Laboratory Animal Sciences, Jinan University; Guangzhou Key Laboratory of Germ-free Animals and Microbiota Application, Guangzhou 510632, China
| | - Jiali Yuan
- School of Basic Medicine, Yunnan University of Traditional Chinese Medicine, China
| | - Qiang Wei
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, China
| | - Mengtao Li
- Department of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences & Peking Union Medical College; Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing 100730, China.
| | - Liangjing Lu
- Department of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200001, China.
| | - Haitao Niu
- School of Medicine and Institute of Laboratory Animal Sciences, Jinan University; Guangzhou Key Laboratory of Germ-free Animals and Microbiota Application, Guangzhou 510632, China; Department of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200001, China.
| |
Collapse
|
77
|
Ha E, Bae SC, Kim K. Recent advances in understanding the genetic basis of systemic lupus erythematosus. Semin Immunopathol 2021; 44:29-46. [PMID: 34731289 DOI: 10.1007/s00281-021-00900-w] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 10/14/2021] [Indexed: 12/22/2022]
Abstract
Systemic lupus erythematosus (SLE) is a polygenic chronic autoimmune disease leading to multiple organ damage. A large heritability of up to 66% is estimated in SLE, with roughly 180 reported susceptibility loci that have been identified mostly by genome-wide association studies (GWASs) and account for approximately 30% of genetic heritability. A vast majority of risk variants reside in non-coding regions, which makes it quite challenging to interpret their functional implications in the SLE-affected immune system, suggesting the importance of understanding cell type-specific epigenetic regulation around SLE GWAS variants. The latest genetic studies have been highly fruitful as several dozens of SLE loci were newly discovered in the last few years and many loci have come to be understood in systemic approaches integrating GWAS signals with other biological resources. In this review, we summarize SLE-associated genetic variants in both the major histocompatibility complex (MHC) and non-MHC loci, examining polygenetic risk scores for SLE and their associations with clinical features. Finally, variant-driven pathogenetic functions underlying genetic associations are described, coupled with discussion about challenges and future directions in genetic studies on SLE.
Collapse
Affiliation(s)
- Eunji Ha
- Department of Biology, Kyung Hee University, Seoul, Republic of Korea.,Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, Seoul, Republic of Korea
| | - Sang-Cheol Bae
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases, Seoul, Republic of Korea. .,Hanyang University Institute for Rheumatology Research, Seoul, Republic of Korea.
| | - Kwangwoo Kim
- Department of Biology, Kyung Hee University, Seoul, Republic of Korea. .,Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, Seoul, Republic of Korea.
| |
Collapse
|
78
|
Xiang K, Wang P, Xu Z, Hu YQ, He YS, Chen Y, Feng YT, Yin KJ, Huang JX, Wang J, Wu ZD, Yang XK, Wang DG, Ye DQ, Pan HF. Causal Effects of Gut Microbiome on Systemic Lupus Erythematosus: A Two-Sample Mendelian Randomization Study. Front Immunol 2021; 12:667097. [PMID: 34557183 PMCID: PMC8453215 DOI: 10.3389/fimmu.2021.667097] [Citation(s) in RCA: 119] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 08/16/2021] [Indexed: 12/19/2022] Open
Abstract
The observational association between gut microbiome and systemic lupus erythematosus (SLE) has been well documented. However, whether the association is causal remains unclear. The present study used publicly available genome-wide association study (GWAS) summary data to perform two-sample Mendelian randomization (MR), aiming to examine the causal links between gut microbiome and SLE. Two sets of MR analyses were conducted. A group of single nucleotide polymorphisms (SNPs) that less than the genome-wide statistical significance threshold (5 × 10-8) served as instrumental variables. To obtain a comprehensive conclusion, the other group where SNPs were smaller than the locus-wide significance level (1 × 10-5) were selected as instrumental variables. Based on the locus-wide significance level, the results indicated that there were causal effects of gut microbiome components on SLE risk. The inverse variance weighted (IVW) method suggested that Bacilli and Lactobacillales were positively correlated with the risk of SLE and Bacillales, Coprobacter and Lachnospira were negatively correlated with SLE risk. The results of weighted median method supported that Bacilli, Lactobacillales, and Eggerthella were risk factors for SLE and Bacillales and Coprobacter served as protective factors for SLE. The estimates of MR Egger suggested that genetically predicted Ruminiclostridium6 was negatively associated with SLE. Based on the genome-wide statistical significance threshold, the results showed that Actinobacteria might reduce the SLE risk. However, Mendelian randomization pleiotropy residual sum and outlier (MR-PRESSO) detected significant horizontal pleiotropy between the instrumental variables of Ruminiclostridium6 and outcome. This study support that there are beneficial or detrimental causal effects of gut microbiome components on SLE risk.
Collapse
Affiliation(s)
- Kun Xiang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China
| | - Peng Wang
- Center for Genetic Epidemiology and Genomics, School of Public Health, Soochow University Medical College, Suzhou, China
| | - Zhiwei Xu
- School of Public Health, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Yu-Qian Hu
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China
| | - Yi-Sheng He
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China
| | - Yue Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China
| | - Ya-Ting Feng
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China
| | - Kang-Jia Yin
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China
| | - Ji-Xiang Huang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China
| | - Jie Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China
| | - Zheng-Dong Wu
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China
| | - Xiao-Ke Yang
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - De-Guang Wang
- Department of Nephrology, Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Dong-Qing Ye
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China
| | - Hai-Feng Pan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China
| |
Collapse
|
79
|
Maglione A, Zuccalà M, Tosi M, Clerico M, Rolla S. Host Genetics and Gut Microbiome: Perspectives for Multiple Sclerosis. Genes (Basel) 2021; 12:1181. [PMID: 34440354 PMCID: PMC8394267 DOI: 10.3390/genes12081181] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/28/2021] [Accepted: 07/28/2021] [Indexed: 12/12/2022] Open
Abstract
As a complex disease, Multiple Sclerosis (MS)'s etiology is determined by both genetic and environmental factors. In the last decade, the gut microbiome has emerged as an important environmental factor, but its interaction with host genetics is still unknown. In this review, we focus on these dual aspects of MS pathogenesis: we describe the current knowledge on genetic factors related to MS, based on genome-wide association studies, and then illustrate the interactions between the immune system, gut microbiome and central nervous system in MS, summarizing the evidence available from Experimental Autoimmune Encephalomyelitis mouse models and studies in patients. Finally, as the understanding of influence of host genetics on the gut microbiome composition in MS is in its infancy, we explore this issue based on the evidence currently available from other autoimmune diseases that share with MS the interplay of genetic with environmental factors (Inflammatory Bowel Disease, Rheumatoid Arthritis and Systemic Lupus Erythematosus), and discuss avenues for future research.
Collapse
Affiliation(s)
- Alessandro Maglione
- Department of Clinical and Biological Sciences, University of Torino, 10100 Torino, Italy; (A.M.); (M.C.)
| | - Miriam Zuccalà
- Department of Health Sciences, Center on Autoimmune and Allergic Diseases (CAAD), Università del Piemonte Orientale, 28100 Novara, Italy; (M.Z.); (M.T.)
| | - Martina Tosi
- Department of Health Sciences, Center on Autoimmune and Allergic Diseases (CAAD), Università del Piemonte Orientale, 28100 Novara, Italy; (M.Z.); (M.T.)
| | - Marinella Clerico
- Department of Clinical and Biological Sciences, University of Torino, 10100 Torino, Italy; (A.M.); (M.C.)
| | - Simona Rolla
- Department of Clinical and Biological Sciences, University of Torino, 10100 Torino, Italy; (A.M.); (M.C.)
| |
Collapse
|
80
|
Levin D, De Palma G, Zou H, Bazzaz AHZ, Verdu E, Baker B, Pinto-Sanchez MI, Khalidi N, Larché MJ, Beattie KA, Bercik P. Fecal microbiome differs between patients with systemic sclerosis with and without small intestinal bacterial overgrowth. JOURNAL OF SCLERODERMA AND RELATED DISORDERS 2021; 6:290-298. [PMID: 35382497 PMCID: PMC8922657 DOI: 10.1177/23971983211032808] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 06/24/2021] [Indexed: 01/04/2023]
Abstract
Introduction: Gastrointestinal manifestations of systemic sclerosis affect up to 90% of
patients, with symptoms including diarrhea and constipation. Small
intestinal bacterial overgrowth is a condition associated with increased
numbers of pathogenic bacteria in the small bowel. While currently unknown,
it has been suggested that dysregulation of the fecal microbiota may play a
role in the development of systemic sclerosis and small intestinal bacterial
overgrowth. Objectives: Our study aimed to describe the fecal microbiota of patients with systemic
sclerosis and compare it between those with and without a diagnosis of small
intestinal bacterial overgrowth. We also compared the fecal microbiota of
systemic sclerosis patients with that of healthy controls to understand the
association between particular bacterial taxa and clinical gastrointestinal
manifestations of systemic sclerosis. Methods: A total of 29 patients with systemic sclerosis underwent breath testing to
assess for small intestinal bacterial overgrowth, provided stool samples to
determine taxonomic assignments, and completed the University of California
Los Angeles Scleroderma Clinical Trial Consortium Gastrointestinal Tract
2.0, which details symptoms and quality-of-life factors. Stool samples were
compared between systemic sclerosis patients with and without small
intestinal bacterial overgrowth, and between patients with systemic
sclerosis and a healthy control cohort (n = 20), aged 18–80 years. Results: Fecal microbiome analyses demonstrated differences between systemic sclerosis
patients with and without small intestinal bacterial overgrowth and
differences in the diversity of species between healthy controls and
patients with systemic sclerosis. Trends were also observed in
anticentromere antibody systemic sclerosis patients, including higher
Alistipies indistincus spp. levels associated with
increased methane levels of breath gas testing and higher
Slakia spp. levels associated with increased rates of
fecal soiling. Conclusions: Our results suggest that changes to the fecal microbiome occur in patients
with small intestinal bacterial overgrowth and systemic sclerosis when
compared to healthy controls. As a cross-sectional study, the potential
pathophysiologic role of an altered microbiome in the development of
systemic sclerosis was not considered and hence needs to be further
investigated.
Collapse
Affiliation(s)
- Daniel Levin
- Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Giada De Palma
- Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Hannah Zou
- Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | | | - Elena Verdu
- Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Barbara Baker
- Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | | | - Nader Khalidi
- Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Maggie J Larché
- Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Karen A. Beattie
- Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Premysl Bercik
- Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
81
|
Correlation Analysis between Gut Microbiota and Metabolites in Children with Systemic Lupus Erythematosus. J Immunol Res 2021; 2021:5579608. [PMID: 34341764 PMCID: PMC8325587 DOI: 10.1155/2021/5579608] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 05/30/2021] [Accepted: 07/02/2021] [Indexed: 12/14/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune-mediated diffuse connective tissue disease characterized by immune inflammation with an unclear aetiology and pathogenesis. This work profiled the intestinal flora and faecal metabolome of patients with SLE using 16S RNA sequencing and gas chromatography-mass spectrometry (GC-MS). We identified unchanged alpha diversity and partially altered beta diversity of the intestinal flora. Another important finding was the increase in Proteobacteria and Enterobacteriales and the decrease in Ruminococcaceae among SLE patients. For metabolites, amino acids and short-chain fatty acids were enriched when long-chain fatty acids were downregulated in SLE faecal samples. KEGG analysis showed the significance of the protein digestion and absorption pathway, and association analysis revealed the key role of 3-phenylpropanoic acid and Sphingomonas. Sphingomonas were reported to be less abundant in healthy periodontal sites of SLE patients than in those of HCs, indicating transmission of oral species to the gut. This study contributes to the understanding of the pathogenesis of SLE disease from the perspective of intestinal microorganisms, explains the pathogenesis of SLE, and serves as a basis for exploring potential treatments for the disease.
Collapse
|
82
|
Altered Profile of Fecal Microbiota in Newly Diagnosed Systemic Lupus Erythematosus Egyptian Patients. Int J Microbiol 2021; 2021:9934533. [PMID: 34257666 PMCID: PMC8249152 DOI: 10.1155/2021/9934533] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 05/16/2021] [Accepted: 06/15/2021] [Indexed: 12/12/2022] Open
Abstract
Background Dysbiosis of gut microbiota could promote autoimmune disorders including systemic lupus erythematosus (SLE). Clarifying this point would be of great importance in understanding the pathogenesis and hence the development of new strategies for SLE treatment. Aim This study aimed to determine the fecal microbiota profile in newly diagnosed SLE patients compared to healthy subjects and to investigate the correlation of this profile with disease activity. Methods Newly diagnosed SLE patients who fulfilled at least four of the American College of Rheumatology (ACR) criteria were enrolled during the study period. Patients with lupus were matched to healthy subjects. SLE activity was evaluated using the Systemic Lupus Disease Activity Index (SLEDAI-2K). Fresh fecal samples were collected from each subject. Genomic DNA was extracted from fecal samples. Quantitative real-time PCR was applied for quantitation of Firmicutes phylum, Bacteroidetes phylum, and Lactobacillus genus in comparison to the total fecal microbiota. Results of patients' samples were compared to those of healthy subjects and were correlated to patients' SLEDAI-2K score. Results Twenty SLE patients' samples were compared with 20 control samples. There was a significant alteration in SLE patients' gut microbiota. A significantly lower (p ≤ 0.001) Firmicutes/Bacteroidetes (F/B) ratio in SLE patients (mean ratio: 0.66%) compared to healthy subjects (mean ratio: 1.79%) was found. Lactobacillus showed a significant decrease in SLE patients (p=0.006) in comparison to healthy controls. An inverse significant correlation between SLEDAI-2K scores for disease activity and F/B ratio (r = -0.451; p=0.04) was found. However, an inverse nonsignificant correlation between SLEDAI-2K scores for disease activity and Lactobacillus (r = -0.155; p=0.51) was detected. Conclusion Compared to healthy controls, recently diagnosed SLE Egyptian patients have an altered fecal microbiota profile with significant lowering of both F/B ratio and Lactobacillus abundance, which is weakly correlated with disease activity.
Collapse
|
83
|
Zhang L, Qing P, Yang H, Wu Y, Liu Y, Luo Y. Gut Microbiome and Metabolites in Systemic Lupus Erythematosus: Link, Mechanisms and Intervention. Front Immunol 2021; 12:686501. [PMID: 34335588 PMCID: PMC8319742 DOI: 10.3389/fimmu.2021.686501] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 06/21/2021] [Indexed: 02/05/2023] Open
Abstract
Systemic lupus erythematosus (SLE), often considered the prototype of autoimmune diseases, is characterized by over-activation of the autoimmune system with abnormal functions of innate and adaptive immune cells and the production of a large number of autoantibodies against nuclear components. Given the highly complex and heterogeneous nature of SLE, the pathogenesis of this disease remains incompletely understood and is presumed to involve both genetic and environmental factors. Currently, disturbance of the gut microbiota has emerged as a novel player involved in the pathogenesis of SLE. With in-depth research, the understanding of the intestinal bacteria-host interaction in SLE is much more comprehensive. Recent years have also seen an increase in metabolomics studies in SLE with the attempt to identify potential biomarkers for diagnosis or disease activity monitoring. An intricate relationship between gut microbiome changes and metabolic alterations could help explain the mechanisms by which gut bacteria play roles in the pathogenesis of SLE. Here, we review the role of microbiota dysbiosis in the aetiology of SLE and how intestinal microbiota interact with the host metabolism axis. A proposed treatment strategy for SLE based on gut microbiome (GM) regulation is also discussed in this review. Increasing our understanding of gut microbiota and their function in lupus will provide us with novel opportunities to develop effective and precise diagnostic strategies and to explore potential microbiota-based treatments for patients with lupus.
Collapse
Affiliation(s)
- Lingshu Zhang
- Department of Rheumatology and Immunology, Rare Diseases Center, Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Pingying Qing
- Department of Rheumatology and Immunology, Rare Diseases Center, Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Hang Yang
- Department of Rheumatology and Immunology, Rare Diseases Center, Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yongkang Wu
- Department of Laboratory Medicine and Outpatient, West China Hospital, Sichuan University, Chengdu, China
| | - Yi Liu
- Department of Rheumatology and Immunology, Rare Diseases Center, Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yubin Luo
- Department of Rheumatology and Immunology, Rare Diseases Center, Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
84
|
Kim JC, Park MJ, Park S, Lee ES. Alteration of the Fecal but Not Salivary Microbiome in Patients with Behçet's Disease According to Disease Activity Shift. Microorganisms 2021; 9:microorganisms9071449. [PMID: 34361885 PMCID: PMC8306658 DOI: 10.3390/microorganisms9071449] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 06/27/2021] [Accepted: 06/29/2021] [Indexed: 12/24/2022] Open
Abstract
The human microbiome plays an important role in various diseases, including Behçet’s disease (BD). However, the effects of disease activity and covariates influencing the microbial composition have not yet been investigated. Therefore, we investigated the fecal and salivary microbiomes of BD patients compared to those of recurrent aphthous ulcer (RAU) patients, as well as dietary habit-matched healthy controls (HCs) selected from immediate family members using 16S rRNA gene sequencing. The fecal microbiome alpha diversity of BD patients was not different from that of their matched HCs, although it was higher than that of unrelated HCs and decreased in BD patients with disease activity. A tendency toward clustering in the beta diversity of the fecal microbiome was observed between the active BD patients and their matched HCs. Active BD patients had a significantly higher abundance of fecal Bacteroides uniformis than their matched HCs and patients with the disease in an inactive state (p = 0.038). The abundance of salivary Rothia mucilaginosa group was higher in BD patients than in RAUs patients. BD patients with uveitis had different abundances of various taxa, compared to those without uveitis. Our results showed an association of fecal microbiome composition with BD disease activity and symptoms, suggesting the possible role of the gut microbiome in BD pathogenesis.
Collapse
Affiliation(s)
- Jin Cheol Kim
- Department of Dermatology, Ajou University School of Medicine, Suwon 16499, Korea; (J.C.K.); (M.J.P.)
| | - Mi Jin Park
- Department of Dermatology, Ajou University School of Medicine, Suwon 16499, Korea; (J.C.K.); (M.J.P.)
| | - Sun Park
- Department of Microbiology and Immunology, Ajou University School of Medicine, Suwon 16499, Korea;
| | - Eun-So Lee
- Department of Dermatology, Ajou University School of Medicine, Suwon 16499, Korea; (J.C.K.); (M.J.P.)
- Correspondence:
| |
Collapse
|
85
|
Vieira JRP, Rezende ATDO, Fernandes MR, da Silva NA. Intestinal microbiota and active systemic lupus erythematosus: a systematic review. Adv Rheumatol 2021; 61:42. [PMID: 34215348 DOI: 10.1186/s42358-021-00201-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 06/20/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Systemic Lupus Erythematosus (SLE) is an autoimmune disease, characterized by being multi-systemic and, therefore, reaching various organs and affecting mainly young women. Its pathogenesis comprehends many factors, including the interaction between microbiota and immune system. This systematic review assessed the relationship between intestinal microbiota and SLE in activity, highlighting microbiota representative patterns regarding quantity and diversity. METHODS This study considered researches carried out in patients with SLE, with no restriction of age or gender, which fulfilled the classification criteria of either Systemic Lupus International Collaborating Clinic (SLICC), American College of Rheumatology (ACR) or European League Against Rheumatism (EULAR) and used the Systemic Lupus Erythematosus Disease Activity Index (SLEDAI) to classify disease in activity or remission were included. The search was carried out from October, 2020 to January, 2021 using the following databases: Medline via Pubmed, Scopus, and Embase. Five papers were included with a total of 288 participants with SLE. RESULTS Regarding microbiota in patients with SLE in activity, there was significant increase in the following genera: Lactobacillus, Streptococcus, Megasphaera, Fusobacterium, Veillonella, Oribacterium, Odoribacter, Blautia, and Campylobacter. On the other hand, decrease in Faecalibacterium and Roseburia genera as well as Ruminococcus gnavus species was observed in remission cases, showing differences between the microbiota profile in SLE in activity and in remission. CONCLUSIONS Results suggest that dysbiosis may be involved in the disease activity process. TRIAL REGISTRATION CRD42021229322 .
Collapse
Affiliation(s)
- Juliana Rosa Pires Vieira
- Postgraduate Program in Health Sciences, School of Medicine of the Universidade Federal de Goiás (UFG), Goiânia, Brazil
| | | | - Marcos Rassi Fernandes
- Postgraduate Program in Health Sciences, School of Medicine of the Universidade Federal de Goiás (UFG), Goiânia, Brazil.,Department of Orthopedics/Traumatology, School of Medicine of the Universidade Federal de Goiás (UFG), Goiânia, Brazil
| | - Nilzio Antonio da Silva
- Postgraduate Program in Health Sciences, School of Medicine of the Universidade Federal de Goiás (UFG), Goiânia, Brazil.,Rheumatology Service of Hospital das Clínicas, School of Medicine of the Universidade Federal de Goiás (UFG), Goiânia, Brazil
| |
Collapse
|
86
|
Liu F, Ren T, Li X, Zhai Q, Xu X, Zhang N, Jiang P, Niu Y, Lv L, Shi G, Feng N. Distinct Microbiomes of Gut and Saliva in Patients With Systemic Lupus Erythematous and Clinical Associations. Front Immunol 2021; 12:626217. [PMID: 34276643 PMCID: PMC8281017 DOI: 10.3389/fimmu.2021.626217] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 06/14/2021] [Indexed: 01/04/2023] Open
Abstract
Alterations in the microbiome of the gut and oral cavity are involved in the etiopathogenesis of systemic lupus erythematosus (SLE). We aimed to assess whether both microbiome compositions in feces and saliva were specific in patients with SLE. A total of 35 patients with SLE, as well as sex- and age-matched asymptomatic subjects as healthy control (HC) group were recruited. Fecal swabs and saliva samples were collected from the participants. 16S ribosomal RNA gene sequencing was performed on the samples. Compared with the HC group, reduced bacterial richness and diversity were detected in the feces of patients with SLE, and increased bacterial diversity in their saliva. Both feces and saliva samples explained the cohort variation. The feces were characterized by enrichment of Lactobacillus, and depletion of an unclassified bacterium in the Ruminococcaceae family and Bifidobacterium. Lack of Bifidobacterium was observed in patients with arthritis. Akkermansia and Ruminococcus negatively correlated with the serum levels of C3. In saliva, Veillonella, Streptococcus, and Prevotella were dominant, and Bacteroides was negatively associated with disease activity. These findings can assist us to comprehensively understand the bacterial profiles of different body niches in SLE patients.
Collapse
Affiliation(s)
- Fengping Liu
- Wuxi School of Medicine, Jiangnan University, Wuxi, China.,Department of Urology, Affiliated Wuxi No. 2 Hospital, Nanjing Medical University, Wuxi, China
| | - Tianli Ren
- Department of Rheumatology, Affiliated Wuxi No. 2 Hospital, Nanjing Medical University, Wuxi, China
| | - Xiaodi Li
- Department of Rheumatology, Affiliated Wuxi No. 2 Hospital, Nanjing Medical University, Wuxi, China
| | - Qixiao Zhai
- State Key Laboratory of Food Science and Technology and School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Xifeng Xu
- Department of Outpatient, Wuxi Children's Hospital of Nanjing Medical University, Wuxi, China
| | - Nan Zhang
- Department of Urology, Affiliated Wuxi No. 2 Hospital, Nanjing Medical University, Wuxi, China
| | - Peng Jiang
- Department of Urology, Affiliated Wuxi No. 2 Hospital, Nanjing Medical University, Wuxi, China
| | - Yaofang Niu
- Research and Development Department, Hangzhou Guhe Information and Technology Company, Hangzhou, China
| | - Longxian Lv
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - GuoXun Shi
- Department of Rheumatology, Affiliated Wuxi No. 2 Hospital, Nanjing Medical University, Wuxi, China
| | - Ninghan Feng
- Department of Urology, Affiliated Wuxi No. 2 Hospital, Nanjing Medical University, Wuxi, China
| |
Collapse
|
87
|
Zhang Y, Xia G, Nie X, Zeng Y, Chen Y, Qian Y, Chen G, Huang J, Wang C, Zhang C, Huang X, Yang Y, Qiu X, Yang F, Chen J, Hu J. Differences in Manifestations and Gut Microbiota Composition Between Patients With Different Henoch-Schonlein Purpura Phenotypes. Front Cell Infect Microbiol 2021; 11:641997. [PMID: 34277463 PMCID: PMC8281929 DOI: 10.3389/fcimb.2021.641997] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 05/12/2021] [Indexed: 01/08/2023] Open
Abstract
Background Gut microbiota plays an important role in the pathogenesis of immune-mediated diseases. However, the complex pathogenesis of Henoch-Schonlein Purpura (HSP) remains elusive. This study aimed to characterize the gut microbiota in HSP patients and explore the potential association between gut microbiota composition and phenotypic changes in HSP. Methods 16SrRNA gene sequencing and bioinformatic analyses were performed using total DNA extracted from the fecal microbiota of 34 children with HSP, including 18 primary cases, 16 recurrent cases, and 23 healthy children. Results The diversity indexes showed significant differences in the microbial community among the primary HSP groups, the recurrent HSP group and healthy controls. The abundance of Escherichia-Shigella in the recurrent HSP group was significantly higher than that in the primary HSP group, and the constructed ROC curve had an AUC value of 0.750. According to the Spearman correlation analysis, the abundance of Bacteroides was positively associated with the serum IgG level in children with HSP, while the abundance of Lachnoclostridium was negatively correlated with the complement component 3 (C3). The diversity indexes of gut microbiota in the HSP group with abdominal symptoms were higher than those in the HSP group without GI involvement, and also higher than those in the healthy control group. In the HSP group with GI involvement, the abundance of Faecalibacterium was decreased, while the abundance of Streptococcus and Fusobacteria was increased, compared to the HSP group without GI involvement. Conclusions The gut microbiota of children with HSP was different from that of healthy children. The genus Escherichia-Shigella has a diagnostic value for HSP recurrence. Bacteroides and Lachnoclostridium may affect IgG and complement C3 levels in children with HSP. Abdominal symptoms in HSP children were related to gut microbiota (Streptococcus and butyric acid-producing bacteria).
Collapse
Affiliation(s)
- Yuanzhen Zhang
- Department of Pediatrics, Affiliated Dongfang Hospital, Xiamen University, Fuzhou, China.,Department of Pediatrics, The 900th Hospital of Joint Logistic Support Force, Fuzhou, China
| | - Guizhi Xia
- Department of Pediatrics, The 900th Hospital of Joint Logistic Support Force, Fuzhou, China.,Department of Pediatrics, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Xiaojing Nie
- Department of Pediatrics, Affiliated Dongfang Hospital, Xiamen University, Fuzhou, China.,Department of Pediatrics, The 900th Hospital of Joint Logistic Support Force, Fuzhou, China.,Department of Pediatrics, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Yugui Zeng
- Department of Pediatrics, Affiliated Dongfang Hospital, Xiamen University, Fuzhou, China
| | - Yi Chen
- Department of Pediatrics, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Yifang Qian
- Department of Pediatrics, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Guangming Chen
- Department of Pediatrics, The 900th Hospital of Joint Logistic Support Force, Fuzhou, China.,Department of Pediatrics, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Jun Huang
- Department of Pediatrics, The 900th Hospital of Joint Logistic Support Force, Fuzhou, China.,Department of Pediatrics, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Chengfeng Wang
- Department of Pediatrics, The 900th Hospital of Joint Logistic Support Force, Fuzhou, China.,Department of Pediatrics, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Chuanyin Zhang
- Department of Pediatrics, The 900th Hospital of Joint Logistic Support Force, Fuzhou, China
| | - Xiaoli Huang
- Department of Pediatrics, The 900th Hospital of Joint Logistic Support Force, Fuzhou, China
| | - Yuen Yang
- Department of Pediatrics, The 900th Hospital of Joint Logistic Support Force, Fuzhou, China
| | - Xiaojian Qiu
- Department of Pediatrics, The Fujian Provincial Maternity and Children's Hospital, Fuzhou, China
| | - Fang Yang
- Department of Pediatrics, Fujian Provincial Hospital, Fuzhou, China
| | - Jie Chen
- Department of Pediatrics, Fujian Provincial Hospital, Fuzhou, China
| | - Jun Hu
- Department of Pediatrics, Fujian Medical University Union Hospital, Fuzhou, China
| |
Collapse
|
88
|
Wu J, Zhuo Y, Liu Y, Chen Y, Ning Y, Yao J. Association between premature ovarian insufficiency and gut microbiota. BMC Pregnancy Childbirth 2021; 21:418. [PMID: 34090383 PMCID: PMC8180047 DOI: 10.1186/s12884-021-03855-w] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 05/05/2021] [Indexed: 01/22/2023] Open
Abstract
Background Premature ovarian insufficiency (POI) is characterized by impairment of ovarian function on a continuum before the age of 40 years. POI is affected by multiple factors. Considering new insights from recent gut microbiome studies, this study aimed to investigate the relationship between gut microbial community structure and POI. Methods Subjects were recruited at the Shenzhen Maternity & Child Healthcare Hospital. Fecal microbial community profiles of healthy women (n = 18), women with POI (n = 35) were analyzed using 16S rRNA gene sequencing based on Illumina NovaSeq platform. Results Compared to the controls, the serum levels of FSH, LH, T and FSH/LH ratio significantly increased in women with POI, whereas E2 and AMH decreased significantly. Higher weighted UniFrac value was observed in POI women compared with healthy women. Phylum Firmicutes, genera Bulleidia and Faecalibacterium were more abundant in healthy women, while phylum Bacteroidetes, genera Butyricimonas, Dorea, Lachnobacterium and Sutterella enriched significantly in women with POI. Moreover, these alterations of the gut microbiome in women with POI were closely related to FSH, LH, E2, AMH level and FSH/LH ratio. Conclusions Women with POI had altered microbial profiles in their gut microbiome, which were associated with serum hormones levels. These results will shed a new light on the pathogenesis and treatment for POI.
Collapse
Affiliation(s)
- Jiaman Wu
- Affiliated Shenzhen Maternity&Child Healthcare Hospital, Southern Medical University, No. 3012, Fuqiang Road, Futian District, Shenzhen City, 518000, Guangdong Province, China
| | - Yuanyuan Zhuo
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518033, China
| | - Yulei Liu
- Affiliated Shenzhen Maternity&Child Healthcare Hospital, Southern Medical University, No. 3012, Fuqiang Road, Futian District, Shenzhen City, 518000, Guangdong Province, China
| | - Yan Chen
- Affiliated Shenzhen Maternity&Child Healthcare Hospital, Southern Medical University, No. 3012, Fuqiang Road, Futian District, Shenzhen City, 518000, Guangdong Province, China
| | - Yan Ning
- Affiliated Shenzhen Maternity&Child Healthcare Hospital, Southern Medical University, No. 3012, Fuqiang Road, Futian District, Shenzhen City, 518000, Guangdong Province, China.
| | - Jilong Yao
- Affiliated Shenzhen Maternity&Child Healthcare Hospital, Southern Medical University, No. 3012, Fuqiang Road, Futian District, Shenzhen City, 518000, Guangdong Province, China.
| |
Collapse
|
89
|
The Immunomodulatory Effect of the Gut Microbiota in Kidney Disease. J Immunol Res 2021; 2021:5516035. [PMID: 34095319 PMCID: PMC8140847 DOI: 10.1155/2021/5516035] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 03/26/2021] [Accepted: 04/20/2021] [Indexed: 12/19/2022] Open
Abstract
The human gut microbiota is a complex cluster composed of 100 trillion microorganisms, which holds a symbiotic relationship with the host under normal circumstances. Intestinal flora can facilitate the treatment of human metabolic dysfunctions and interact with the intestinal tract, which could influence intestinal tolerance, immunity, and sensitivity to inflammation. In recent years, significant interests have evolved on the association of intestinal microbiota and kidney diseases within the academic circle. Abnormal changes in intestinal microbiota, known as dysbiosis, can affect the integrity of the intestinal barrier, resulting in the bacterial translocation, production, and accumulation of dysbiotic gut-derived metabolites, such as urea, indoxyl sulfate (IS), and p-cresyl sulfate (PCS). These processes lead to the abnormal activation of immune cells; overproduction of antibodies, immune complexes, and inflammatory factors; and inflammatory cell infiltration that can directly or indirectly cause damage to the renal parenchyma. The aim of this review is to summarize the role of intestinal flora in the development and progression of several renal diseases, such as lupus nephritis, chronic kidney disease, diabetic nephropathy, and renal ischemia-reperfusion injury. Further research on these mechanisms should provide insights into the therapeutic potential of regulating intestinal flora and intervening related molecular targets for the abovementioned nephropathy.
Collapse
|
90
|
Wei M, Li C, Dai Y, Zhou H, Cui Y, Zeng Y, Huang Q, Wang Q. High-Throughput Absolute Quantification Sequencing Revealed Osteoporosis-Related Gut Microbiota Alterations in Han Chinese Elderly. Front Cell Infect Microbiol 2021; 11:630372. [PMID: 33996619 PMCID: PMC8120270 DOI: 10.3389/fcimb.2021.630372] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 04/19/2021] [Indexed: 12/06/2022] Open
Abstract
Objective Accumulative evidence suggests that gut microbiota play an important role in bone remodeling and hence bone health maintenance. This study aimed to explore the association of gut microbiota with the risk of osteoporosis and to identify potential disease-related taxa, which may be promising targets in osteoporosis prevention and treatment in the future. Methods Absolute quantification 16S ribosomal RNA gene sequencing was used to detect absolute and relative abundances of gut microbiota in 44 patients with osteoporosis and 64 controls. In combination with one of our previous studies, a total of 175 samples were involved in the relative abundance analysis. Results Compared with the controls, the patients with osteoporosis had higher absolute and relative abundances of Bacteroidetes phylum, and Bacteroides and Eisenbergiella genera. The absolute abundances of Clostridium_XlVa, Coprococcus, Lactobacillus, and Eggerthella genera increased, and that of the Veillonella genus decreased in the osteoporosis group. As for relative abundance, that of the Parabacteroides and Flavonifractor genera increased, whereas that of the Raoultella genus decreased in the osteoporosis group. Controlling for potential confounders, the associations of Clostridium_XlVa, Coprococcus, and Veillonella genera with the risk of osteoporosis did not maintain significance. Ridge regression analysis suggested that Bacteroides is associated with reduced bone mineral density (BMD) and T-score at lumbar spines, and Anaerovorax is associated with increased BMD at the femoral neck. Functional predictions revealed that 10 Kyoto Encyclopedia of Genes and Genomes pathways were enriched in the osteoporosis group. Conclusions Gut microbiota compositions may contribute to the risk of osteoporosis. Several specific taxa and functional pathways are identified to associate with reduced bone density, thus providing epidemiologic evidence for the potential role of aberrant gut microbiota in osteoporosis pathogenesis.
Collapse
Affiliation(s)
- Muhong Wei
- MOE Key Lab of Environment and Health, Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Can Li
- MOE Key Lab of Environment and Health, Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Dai
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haolong Zhou
- MOE Key Lab of Environment and Health, Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuan Cui
- MOE Key Lab of Environment and Health, Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yun Zeng
- Department of Medical Record Statistics, Wuhan NO.1 Hospital, Wuhan, China
| | - Qin Huang
- Department of Rehabilitation Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qi Wang
- MOE Key Lab of Environment and Health, Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
91
|
Wu X, Chen X, Lyu X, Zheng H. Advances in Microbiome Detection Technologies and Application in Antirheumatic Drug Design. Curr Pharm Des 2021; 27:891-899. [PMID: 33308114 DOI: 10.2174/1381612826666201211114609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 08/25/2020] [Indexed: 11/22/2022]
Abstract
Rheumatic diseases are a kind of chronic inflammatory and autoimmune disease affecting the connection or supporting structures of the human body, such as the most common diseases Ankylosing spondylitis (AS), gout and Systemic lupus erythematosus (SLE). Although the precise etiology and pathogenesis of the different types of rheumatic diseases remain mostly unknown, it is now commonly believed that these diseases are attributed to some complex interactions between genetics and environmental factors, especially the gut microbiome. Altered microbiome showed clinical improvement in disease symptoms and partially restored to normality after prescribing disease-modifying antirheumatic drugs (DMARDs) or other treatment strategies. Recent advances in next-generation sequencing-based microbial profiling technology, especially metagenomics, have identified alteration of the composition and function of the gut microbiota in patients. Clinical and experimental data suggest that dysbiosis may play a pivotal role in the pathogenesis of these diseases. In this paper, we provide a brief review of the advances in the microbial profiling technology and up-to-date resources for accurate taxonomic assignment of metagenomic reads, which is a key step for metagenomics studies. In addition, we review the altered gut microbiota signatures that have been reported so far across various studies, upon which diagnostics classification models can be constructed, and the drug-induced regulation of the host microbiota can be used to control disease progression and symptoms.
Collapse
Affiliation(s)
- Xin Wu
- Department of Rheumatology and Immunology, Shanghai Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai, China
| | - Xiang Chen
- Department of Bioinformatics, Hangzhou Nuowei Information Technology, Co., Ltd. Hangzhou, China
| | - Xiaochen Lyu
- Department of Bioinformatics, Hangzhou Nuowei Information Technology, Co., Ltd. Hangzhou, China
| | - Hao Zheng
- Department of Bioinformatics, Hangzhou Nuowei Information Technology, Co., Ltd. Hangzhou, China
| |
Collapse
|
92
|
Li R, Meng X, Chen B, Zhao L, Zhang X. Gut Microbiota in Lupus: a Butterfly Effect? Curr Rheumatol Rep 2021; 23:27. [PMID: 33864162 DOI: 10.1007/s11926-021-00986-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/19/2021] [Indexed: 12/16/2022]
Abstract
PURPOSE OF REVIEW Systemic lupus erythematosus (SLE) is a prototypic autoimmune disease that typically displays chronic inflammatory tissue damage and miscellaneous circulatory autoantibodies, as well as distinctive type 1 interferon signatures. The etiology of SLE is unclear and currently is attributed to genetic predisposition and environmental triggers. Gut microbiota has recently been considered a critical environmental pathogenic factor in immune-related disorders, and studies are ongoing to uncover the key pathogens and the imputative mechanisms. Fundamental advancements on the role of the microbiota in SLE pathology have been achieved in recent years and are summarized in this review. RECENT FINDINGS Recent findings suggested that gut commensals could propagate autoimmunity via molecular mimicry in which ortholog-carrying microbes cross-activate autoreactive T/B cells and trigger the response against host autoantigens, or via bystander activation by stimulating antigen-presenting cells that present autoantigens and enhancing the expression of co-stimulatory molecules and cytokines, thus leading to the loss of self-tolerance and the production of autoantibodies. Additionally, the break of gut barrier and the translocation of gut commensals to inner organs can trigger immune dysregulation and inappropriate systemic inflammation. All these microbiota-mediated mechanisms could contribute to lupus immunopathogenesis and promote disease development in susceptible individuals. Evidence of the causative role of disturbed gut microbiome in SLE is still limited, and the related molecular mechanisms and pathways are largely elusive. However, the modification of gut microbiota, such as pathobiont vaccine, special diet, restricted consortium transplantation, as well as regulatory metabolites supplementation, might be promising strategies for lupus prophylaxis and treatment.
Collapse
Affiliation(s)
- Rongli Li
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, The Ministry of Education Key Laboratory, Chinese Academy of Medical Sciences and Peking Union Medical College, Shuai-Fu-Yuan, Dong-Cheng District, Beijing, 100730, China
| | - Xia Meng
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, The Ministry of Education Key Laboratory, Chinese Academy of Medical Sciences and Peking Union Medical College, Shuai-Fu-Yuan, Dong-Cheng District, Beijing, 100730, China
| | - Beidi Chen
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, The Ministry of Education Key Laboratory, Chinese Academy of Medical Sciences and Peking Union Medical College, Shuai-Fu-Yuan, Dong-Cheng District, Beijing, 100730, China
| | - Lidan Zhao
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, The Ministry of Education Key Laboratory, Chinese Academy of Medical Sciences and Peking Union Medical College, Shuai-Fu-Yuan, Dong-Cheng District, Beijing, 100730, China. .,National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID); State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (PUMCH), 41 Damucang Hutong, Xicheng District, Beijing, 100730, China.
| | - Xuan Zhang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, The Ministry of Education Key Laboratory, Chinese Academy of Medical Sciences and Peking Union Medical College, Shuai-Fu-Yuan, Dong-Cheng District, Beijing, 100730, China. .,Clinical Immunology Centre, Medical Epigenetics Research Centre, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shuai-Fu-Yuan, Dong-Cheng District, Beijing, 100730, China.
| |
Collapse
|
93
|
Mok WK, Tan YX, Chen WN. Evaluating the potential of Bacillus subtilis fermented okara as a functional food ingredient through in vitro digestion and fermentation. FOOD BIOTECHNOL 2021. [DOI: 10.1080/08905436.2021.1909615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Wai Kit Mok
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, Singapore
| | - Yong Xing Tan
- Interdisciplinary Graduate School, Nanyang Technological University, Singapore, Singapore
- Advanced Environmental Biotechnology Centre, Nanyang Environment and Water Research Institute, Nanyang Technological University, Singapore, Singapore
| | - Wei Ning Chen
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
94
|
Colucci R, Moretti S. Implication of Human Bacterial Gut Microbiota on Immune-Mediated and Autoimmune Dermatological Diseases and Their Comorbidities: A Narrative Review. Dermatol Ther (Heidelb) 2021; 11:363-384. [PMID: 33507493 PMCID: PMC8018919 DOI: 10.1007/s13555-021-00485-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Indexed: 12/12/2022] Open
Abstract
During the last decade, the advent of modern sequencing methods (next generation techniques, NGS) has helped describe the composition of the human gut microbiome, enabling us to understand the main characteristics of a healthy gut microbiome and, conversely, the magnitude of its disease-related changes. This new knowledge has revealed that healthy gut microbiota allow the maintenance of several crucial physiological functions, such as the ability to regulate the innate and adaptive immune systems. Increasing evidence has pointed out a condition of dysbiosis in several autoimmune/immune mediated dermatological conditions and specific gut microbial signatures have also been reported to correlate with clinical and prognostic parameters of such diseases. Based on a literature search of relevant published articles, this review debates the current knowledge and the possible pathogenic implications of bacterial gut microbiota composition assessed through NGS techniques in systemic lupus erythematosus, atopic dermatitis, psoriasis, and alopecia areata. Evidence of a potential role of specific gut microbiota signatures in modulating the clinical course of such diseases and their main comorbidities has been also reviewed.
Collapse
Affiliation(s)
- Roberta Colucci
- Section of Dermatology, Department of Health Sciences, University of Florence, Florence, Italy.
| | - Silvia Moretti
- Section of Dermatology, Department of Health Sciences, University of Florence, Florence, Italy
| |
Collapse
|
95
|
Yu G, Ji X, Huang J, Liao A, Pan L, Hou Y, Hui M, Guo W. Immunity improvement and gut microbiota remodeling of mice by wheat germ globulin. World J Microbiol Biotechnol 2021; 37:64. [PMID: 33733383 DOI: 10.1007/s11274-021-03034-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 03/08/2021] [Indexed: 12/27/2022]
Abstract
The wheat germ protein (WG) and it's proteolytic peptide have a variety of biological activities. Our previous work showed that WG could improve immunity of the immunosuppressive mice established by cyclophosphamide. However, in the healthy condition and normal diet, as a supplementary food, the effects of immunity improvement and gut microbiota remodeling by the wheat germ globulin has not been studied yet. Here, we reported that WG could improve the immunity and remodel the gut microbiota of the mice, as a potentially safe functional supplementary food for the first time. The increase of interleukin-6 (IL-6) and the decrease of tumor necrosis factor α (TNF-α) and interleukin-10 (IL-10) indicated that WG could enhance the levels of activated T cells and monocytes and anti-inflammatory ability, meanwhile, the significant increase of immunoglobin G (lgG) and the notable decrease of the immunoglobin M (lgM) and immunoglobin A (lgA) illustrated that WG could improve immunity by promoting the differentiation and maturation process of B cells, compared with the NC group (normal control group). 16S rRNA sequencing showed WG could remodel the gut microbiota. At the phylum level, the Bacteroidetes were reduced and Firmicutes were increased in WG group, compared with NC group. At the genus level, the SCFA producing genera of unclassified_f_Lachnospiraceae, Blautia and especially the Roseburia (increased more than threefold) increased notably. Further, the level changes of cytokines and immunoglobulins were associated with the gut microbiota. This work showed that WG could improve immunity and has potential application value as an immune-enhancing functional food.
Collapse
Affiliation(s)
- Guanghai Yu
- College of Biological Engineering, Henan University of Technology, Zhengzhou, 450001, P. R. China
- Henan Provincial Key Laboratory of Biological Processing and Nutritional Function of Wheat, Zhengzhou, 450001, P. R. China
| | - Xiaoguo Ji
- College of Biological Engineering, Henan University of Technology, Zhengzhou, 450001, P. R. China
| | - Jihong Huang
- College of Biological Engineering, Henan University of Technology, Zhengzhou, 450001, P. R. China.
- Henan Provincial Key Laboratory of Biological Processing and Nutritional Function of Wheat, Zhengzhou, 450001, P. R. China.
| | - Aimei Liao
- College of Biological Engineering, Henan University of Technology, Zhengzhou, 450001, P. R. China
- Henan Provincial Key Laboratory of Biological Processing and Nutritional Function of Wheat, Zhengzhou, 450001, P. R. China
| | - Long Pan
- College of Biological Engineering, Henan University of Technology, Zhengzhou, 450001, P. R. China
- Henan Provincial Key Laboratory of Biological Processing and Nutritional Function of Wheat, Zhengzhou, 450001, P. R. China
| | - Yinchen Hou
- National Engineering Laboratory for Wheat & Corn Further Processing, Henan University of Technology, Zhengzhou, 450001, P. R. China
- Henan University of Animal Husbandry Economy, Zhengzhou, 450046, P. R. China
| | - Ming Hui
- College of Biological Engineering, Henan University of Technology, Zhengzhou, 450001, P. R. China
- Henan Provincial Key Laboratory of Biological Processing and Nutritional Function of Wheat, Zhengzhou, 450001, P. R. China
| | - Weiyun Guo
- Food and Pharmacy College, Xuchang University, Xuchang, 461000, P. R. China
| |
Collapse
|
96
|
Shi XR, Chen BY, Lin WZ, Li YL, Wang YL, Liu Y, Huang JJ, Zhang WW, Ma XX, Shao S, Li RG, Duan SZ. Microbiota in Gut, Oral Cavity, and Mitral Valves Are Associated With Rheumatic Heart Disease. Front Cell Infect Microbiol 2021; 11:643092. [PMID: 33768014 PMCID: PMC7985333 DOI: 10.3389/fcimb.2021.643092] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 02/04/2021] [Indexed: 12/20/2022] Open
Abstract
Rheumatic heart disease refers to the long-term damage of heart valves and results from an autoimmune response to group A Streptococcus infection. This study aimed to analyze the microbiota composition of patients with rheumatic heart disease and explore potential function of microbiota in this disease. First, we revealed significant alterations of microbiota in feces, subgingival plaques, and saliva of the patients compared to control subjects using 16S rRNA gene sequencing. Significantly different microbial diversity was observed in all three types of samples between the patients and control subjects. In the gut, the patients possessed higher levels of genera including Bifidobacterium and Eubacterium, and lower levels of genera including Lachnospira, Bacteroides, and Faecalibacterium. Coprococcus was identified as a super-generalist in fecal samples of the patients. Significant alterations were also observed in microbiota of subgingival plaques and saliva of the patients compared to control subjects. Second, we analyzed microbiota in mitral valves of the patients and identified microbes that could potentially transmit from the gut or oral cavity to heart valves, including Streptococcus. Third, we further analyzed the data using random forest model and demonstrated that microbiota in the gut, subgingival plaque or saliva could distinguish the patients from control subjects. Finally, we identified gut/oral microbes that significantly correlated with clinical indices of rheumatic heart disease. In conclusion, patients with rheumatic heart disease manifested important alterations in microbiota that might distinguish the patients from control subjects and correlated with severity of this disease.
Collapse
Affiliation(s)
- Xue-Rui Shi
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Bo-Yan Chen
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Wen-Zhen Lin
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Yu-Lin Li
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Yong-Li Wang
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Yan Liu
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Jing-Juan Huang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Wei-Wei Zhang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xiao-Xin Ma
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Shuai Shao
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Ruo-Gu Li
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Sheng-Zhong Duan
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| |
Collapse
|
97
|
Mangalam AK, Yadav M, Yadav R. The Emerging World of Microbiome in Autoimmune Disorders: Opportunities and Challenges. INDIAN JOURNAL OF RHEUMATOLOGY 2021; 16:57-72. [PMID: 34531642 PMCID: PMC8442979 DOI: 10.4103/injr.injr_210_20] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Trillions of commensal bacteria colonizing humans (microbiome) have emerged as essential player(s) in human health. The alteration of the same has been linked with diseases including autoimmune disorders such as multiple sclerosis, rheumatoid arthritis, systemic lupus erythematosus, and ankylosing spondylitis. Gut bacteria are separated from the host through a physical barrier such as skin or gut epithelial lining. However, the perturbation in the healthy bacterial community (gut dysbiosis) can compromise gut barrier integrity, resulting in translocation of bacterial contents across the epithelial barrier (leaky gut). Bacterial contents such as lipopolysaccharide and bacterial antigens can induce a systemic inflammatory environment through activation and induction of immune cells. The biggest question in the field is whether inflammation causes gut dysbiosis or dysbiosis leads to disease induction or propagation, i.e., it is inside out or outside in or both. In this review, we first discuss the microbiome profiling studies in various autoimmune disorders, followed by a discussion of potential mechanisms through which microbiome is involved in the pathobiology of diseases. A better understanding of the role of the microbiome in health and disease will help us harness the power of commensal bacteria for the development of novel therapeutic agents to treat autoimmune disorders.
Collapse
Affiliation(s)
| | - Meeta Yadav
- Department of Pathology, University of Iowa, Iowa, IA,
USA
| | - Rajwardhan Yadav
- Department of Rheumatology, St Francis Hospital, Hartford,
CT, USA
| |
Collapse
|
98
|
Maalouly G, Hajal J, Noujeim C, Choueiry M, Nassereddine H, Smayra V, Saliba Y, Fares N. New insights in gut-liver axis in wild-type murine imiquimod-induced lupus. Lupus 2021; 30:926-936. [PMID: 33596715 DOI: 10.1177/0961203321995254] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND Intestinal and hepatic manifestations of lupus seem to be underestimated in comparison to other major organ lesions. Although recent data point to gut-liver axis involvement in lupus, gut permeability dysfunction and liver inflammation need to be more investigated. OBJECTIVE This study aims to assess fecal calprotectin, intestinal tight junction proteins and liver inflammation pathway in wild-type murine imiquimod- induced lupus. METHODS C57BL/6 mice were topically treated on their right ears with 1.25 mg of 5% imiquimod cream, three times per week for six weeks. Fecal calprotectin was collected at day 0, 22 and 45. Renal, liver and intestinal pathology, as well as inflammatory markers, intestinal tight junction proteins, and E. coli protein in liver were assessed at sacrifice. RESULTS At six weeks, lupus nephritis was confirmed on histopathology and NGAL and KIM-1 expression. Calprotectin rise started at day 22 and persists at day 45. Protein expression of Claudine, ZO-1 and occludin was significantly decreased. E. coli protein was significantly increased in liver with necro-inflammation and increased TLR4, TLR7, and pNFκB/NFκB liver expression. CONCLUSION This study is the first to demonstrate early fecal calprotectin increase and liver activation of TLR4- NFκB pathway in wild-type murine imiquimod-induced lupus.
Collapse
Affiliation(s)
- Georges Maalouly
- Faculty of Medicine, CHU Hotel Dieu de France Hospital, Saint Joseph University, Beirut, Lebanon
| | - Joelle Hajal
- Physiology and Pathophysiology Research Laboratory, Pole of Technology and Health, Faculty of Medicine, Saint Joseph University, Beirut, Lebanon
| | - Charbel Noujeim
- Physiology and Pathophysiology Research Laboratory, Pole of Technology and Health, Faculty of Medicine, Saint Joseph University, Beirut, Lebanon
| | - Michel Choueiry
- Physiology and Pathophysiology Research Laboratory, Pole of Technology and Health, Faculty of Medicine, Saint Joseph University, Beirut, Lebanon
| | - Hussein Nassereddine
- Faculty of Medicine, CHU Hotel Dieu de France Hospital, Saint Joseph University, Beirut, Lebanon
| | - Viviane Smayra
- Faculty of Medicine, CHU Hotel Dieu de France Hospital, Saint Joseph University, Beirut, Lebanon
| | - Youakim Saliba
- Physiology and Pathophysiology Research Laboratory, Pole of Technology and Health, Faculty of Medicine, Saint Joseph University, Beirut, Lebanon
| | - Nassim Fares
- Physiology and Pathophysiology Research Laboratory, Pole of Technology and Health, Faculty of Medicine, Saint Joseph University, Beirut, Lebanon
| |
Collapse
|
99
|
Nanoparticles in the Food Industry and Their Impact on Human Gut Microbiome and Diseases. Int J Mol Sci 2021; 22:ijms22041942. [PMID: 33669290 PMCID: PMC7920074 DOI: 10.3390/ijms22041942] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 02/02/2021] [Accepted: 02/10/2021] [Indexed: 12/11/2022] Open
Abstract
The use of inorganic nanoparticles (NPs) has expanded into various industries including food manufacturing, agriculture, cosmetics, and construction. This has allowed NPs access to the human gastrointestinal tract, yet little is known about how they may impact human health. As the gut microbiome continues to be increasingly implicated in various diseases of unknown etiology, researchers have begun studying the potentially toxic effects of these NPs on the gut microbiome. Unfortunately, conflicting results have limited researcher’s ability to evaluate the true impact of NPs on the gut microbiome in relation to health. This review focuses on the impact of five inorganic NPs (silver, iron oxide, zinc oxide, titanium dioxide, and silicon dioxide) on the gut microbiome and gastrointestinal tract with consideration for various methodological differences within the literature. This is important as NP-induced changes to the gut could lead to various gut-related diseases. These include irritable bowel syndrome (IBS), inflammatory bowel disease (IBD), celiac disease, and colorectal cancer. Research in this area is necessary as the use of NPs in various industries continues to grow along with the number of people suffering from chronic gastrointestinal diseases.
Collapse
|
100
|
Katz-Agranov N, Zandman-Goddard G. The microbiome links between aging and lupus. Autoimmun Rev 2021; 20:102765. [PMID: 33476814 DOI: 10.1016/j.autrev.2021.102765] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 11/05/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND AND AIMS Many forms of immune dysregulation, which lead to inflammaging and senescence, have been demonstrated in patients with systemic lupus erythematosus (SLE; lupus) and in the aging population. The discovery of the microbiome and its association with human health and pathology has led it to be the center of investigation as a major contributor to the pathogenesis of immunosenescence in both populations. Similar alterations to the microbiome in the form of dysbiosis, that are demonstrated in both aging and in lupus patients, may help explain the significant overlap in clinical manifestations seen in these groups. METHODS We performed an extensive literature review, utilizing the Pubmed search engine and Google Scholar for studies evaluating the microbiome in two groups, elderly populations and lupus patients (both murine and human models), between the years 2000-2019. We searched for the terms: microbiome, dysbiosis, lupus, elderly, aging and inflammaging, which yielded hundreds of articles, of which 114 were used for preparation of this paper. We compared the similarities between the populations. RESULTS We found that the similar processes of immune dysregulation, in both aging populations and lupus patients, extend to the microbiome, in the form of dysbiosis. Some of these similarities include loss of microbiota biodiversity, increased representation of microbes that are associated with inflammation and disease (i.e Proteobacteria, Bacteroidetes), a relative decrease in protective microbes with "anti-inflammatory" properties (i.e Firmicutes) and a subsequent compromise to the intestinal barrier, leading to leakage of proinflammatory microbial components in both groups. CONCLUSIONS We conclude that there are several similar alterations in the composition and function of the microbiome of lupus patients and aging individuals, leading to immunosenescence, which may also be a contributing mechanism in lupus. It seems in fact that the microbiome of SLE may actually be analogous to immunosenescence. This knowledge may help the continuous efforts in finding a solution for both conditions.
Collapse
Affiliation(s)
- Nurit Katz-Agranov
- Department of Medicine, Saint Elizabeth's Medical Center, Boston, MA, USA; Tufts University School of Medicine, Boston, MA, USA
| | - Gisele Zandman-Goddard
- Department of Medicine C, Wolfson Medical Center, Holon, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|