Microbiome in Behcet's syndrome.

[1]  G. Hatemi,et al.  Vascular Behçet syndrome: from pathogenesis to treatment , 2022, Nature Reviews Rheumatology.

[2]  Hyon Ju Park,et al.  Behcet disease: an undifferentiating and complex vasculitis , 2022, Postgraduate medicine.

[3]  Z. Zádori,et al.  Interactions between NSAIDs, opioids and the gut microbiota - Future perspectives in the management of inflammation and pain. , 2022, Pharmacology & therapeutics.

[4]  D. Roccatello,et al.  The role of bacteria and viruses in Behçet syndrome: Should we move towards new paradigms? , 2022, Autoimmunity reviews.

[5]  W. Robinson,et al.  Clonal IgA and IgG autoantibodies from individuals at risk for rheumatoid arthritis identify an arthritogenic strain of Subdoligranulum , 2022, Science Translational Medicine.

[6]  A. Kurilshikov,et al.  Gut microbiome dysbiosis is associated with increased mortality after solid organ transplantation , 2022, Science Translational Medicine.

[7]  Anna Christovich,et al.  Gut Microbiota, Leaky Gut, and Autoimmune Diseases , 2022, Frontiers in Immunology.

[8]  Xing He,et al.  Findings on the Relationship Between Intestinal Microbiome and Vasculitis , 2022, Frontiers in Cellular and Infection Microbiology.

[9]  T. Iqbal,et al.  Gut microbiome and autoimmune disorders. , 2022, Clinical and experimental immunology.

[10]  G. Hatemi,et al.  Behçet's syndrome: one year in review 2022. , 2022, Clinical and experimental rheumatology.

[11]  E. Miyauchi,et al.  The impact of the gut microbiome on extra-intestinal autoimmune diseases , 2022, Nature Reviews Immunology.

[12]  D. Mozaffarian,et al.  Association of Trimethylamine N-Oxide and Metabolites With Mortality in Older Adults , 2022, JAMA network open.

[13]  A. Fateh,et al.  Interaction between Clostridium species and microbiota to progress immune regulation. , 2022, Acta microbiologica et immunologica Hungarica.

[14]  D. Ojcius,et al.  Gut barrier disruption and chronic disease , 2022, Trends in Endocrinology & Metabolism.

[15]  A. Arcangeli,et al.  A unique circulating miRNA profile highlights thrombo-inflammation in Behçet’s syndrome , 2021, Annals of the Rheumatic Diseases.

[16]  S. Baldovino,et al.  Vedolizumab for the Management of Refractory Behçet’s Disease: From a Case Report to New Pieces of Mosaic in a Complex Disease , 2021, Frontiers in Immunology.

[17]  H. Direskeneli,et al.  Pathogenesis of Behçet's Syndrome: Genetic, Environmental and Immunological Factors , 2021, Frontiers in Medicine.

[18]  N. Taddei,et al.  Neutrophil‐mediated mechanisms of damage and in‐vitro protective effect of colchicine in non‐vascular Behçet’s syndrome , 2021, Clinical and experimental immunology.

[19]  A. Kijlstra,et al.  Changes in the Gut Microbiome Contribute to the Development of Behcet’s Disease via Adjuvant Effects , 2021, Frontiers in Cell and Developmental Biology.

[20]  C. Suh,et al.  Eubacterium rectale Attenuates HSV-1 Induced Systemic Inflammation in Mice by Inhibiting CD83 , 2021, Frontiers in Immunology.

[21]  D. Mozaffarian,et al.  Association of Trimethylamine N-Oxide and Related Metabolites in Plasma and Incident Type 2 Diabetes , 2021, JAMA network open.

[22]  Sun Park,et al.  Alteration of the Fecal but Not Salivary Microbiome in Patients with Behçet’s Disease According to Disease Activity Shift , 2021, Microorganisms.

[23]  G. Wallace,et al.  Behçet’s Disease—Do Microbiomes and Genetics Collaborate in Pathogenesis? , 2021, Frontiers in Immunology.

[24]  N. Mizuki,et al.  Changes in the proportion of clinical clusters contribute to the phenotypic evolution of Behçet’s disease in Japan , 2021, Arthritis Research & Therapy.

[25]  J. Guan,et al.  Cluster analysis of phenotypes of patients with Behçet’s syndrome: a large cohort study from a referral center in China , 2020, Arthritis Research & Therapy.

[26]  D. Dordević,et al.  Hydrogen sulfide toxicity in the gut environment: Meta-analysis of sulfate-reducing and lactic acid bacteria in inflammatory processes , 2020, Journal of advanced research.

[27]  R. Moots,et al.  A Darwinian View of Behçet's Disease , 2015, Rheumatology and immunology research.

[28]  Samir A. Shah,et al.  Immunoglobulin A Targets a Unique Subset of the Microbiota in Inflammatory Bowel Disease. , 2020, Cell host & microbe.

[29]  F. Sofi,et al.  Butyrate Rich Diets Improve Redox Status and Fibrin Lysis in Behçet's Syndrome. , 2020, Circulation research.

[30]  Ç. Ergin,et al.  Succinivibrionaceae is dominant family in fecal microbiota of Behçet’s Syndrome patients with uveitis , 2020, PloS one.

[31]  A. Moya,et al.  Intestinal microbiota composition of patients with Behçet's disease: differences between eye, mucocutaneous and vascular involvement. The Rheuma-BIOTA study. , 2020, Clinical and experimental rheumatology.

[32]  N. D. de Boer,et al.  Gut Microbiota-driven Drug Metabolism in Inflammatory Bowel Disease , 2020, Journal of Crohn's & colitis.

[33]  Malbert R. C. Rogers,et al.  Behçet's Disease Under Microbiotic Surveillance? A Combined Analysis of Two Cohorts of Behçet's Disease Patients , 2020, Frontiers in Immunology.

[34]  D. Seals,et al.  Trimethylamine-N-Oxide Promotes Age-Related Vascular Oxidative Stress and Endothelial Dysfunction in Mice and Healthy Humans , 2020, Hypertension.

[35]  E. Silvestri,et al.  Long-term effectiveness and safety of secukinumab for treatment of refractory mucosal and articular Behçet’s phenotype: a multicentre study , 2020, Annals of the Rheumatic Diseases.

[36]  P. Turnbaugh,et al.  Pharmacomicrobiomics in inflammatory arthritis: gut microbiome as modulator of therapeutic response , 2020, Nature Reviews Rheumatology.

[37]  Oliver Fiehn,et al.  A Cardiovascular Disease-Linked Gut Microbial Metabolite Acts via Adrenergic Receptors , 2020, Cell.

[38]  M. Osman,et al.  Global disparities in faecal microbiota transplantation research. , 2020, The lancet. Gastroenterology & hepatology.

[39]  S. Turroni,et al.  Modulation of gut microbiota through nutritional interventions in Behçet’s syndrome patients (the MAMBA study): study protocol for a randomized controlled trial , 2019, Trials.

[40]  S. Ohno,et al.  Alteration of oral flora in Mongolian patients with Behçet's disease: a multicentre study. , 2020, Clinical and experimental rheumatology.

[41]  W. Dahl,et al.  Diet, nutrients and the microbiome. , 2020, Progress in molecular biology and translational science.

[42]  Y. Tao,et al.  Demystifying the manipulation of host immunity, metabolism, and extraintestinal tumors by the gut microbiome , 2019, Signal Transduction and Targeted Therapy.

[43]  S. Turner,et al.  Microbiota-Derived Short-Chain Fatty Acids Promote the Memory Potential of Antigen-Activated CD8+ T Cells. , 2019, Immunity.

[44]  Anukriti Sharma,et al.  Pharmacomicrobiomics: The Holy Grail to Variability in Drug Response? , 2019, Clinical pharmacology and therapeutics.

[45]  N. Oezguen,et al.  Microbiota stratification identifies disease-specific alterations in neuro-Behçet's disease and multiple sclerosis. , 2019, Clinical and experimental rheumatology.

[46]  G. Hatemi,et al.  Behçet's Syndrome as a Model of Thrombo-Inflammation: The Role of Neutrophils , 2019, Front. Immunol..

[47]  E. Alpsoy,et al.  Behçet’s Disease: An Overview of Etiopathogenesis , 2019, Front. Immunol..

[48]  A. R. Moravejolahkami,et al.  Synbiotic Supplementation May Relieve Anterior Uveitis, an Ocular Manifestation in Behcet’s Syndrome , 2019, The American journal of case reports.

[49]  Y. Ghasemi,et al.  Prebiotics: Definition, Types, Sources, Mechanisms, and Clinical Applications , 2019, Foods.

[50]  N. Taddei,et al.  Behçet’s syndrome as a tool to dissect the mechanisms of thrombo‐inflammation: clinical and pathogenetic aspects , 2018, Clinical and experimental immunology.

[51]  E. Silvestri,et al.  Efficacy of the anti-IL 17 secukinumab in refractory Behçet's syndrome: A preliminary study. , 2019, Journal of autoimmunity.

[52]  A. Gasbarrini,et al.  What is the Healthy Gut Microbiota Composition? A Changing Ecosystem across Age, Environment, Diet, and Diseases , 2019, Microorganisms.

[53]  J. Shimizu,et al.  Relative abundance of Megamonas hypermegale and Butyrivibrio species decreased in the intestine and its possible association with the T cell aberration by metabolite alteration in patients with Behcet’s disease (210 characters) , 2019, Clinical Rheumatology.

[54]  M. Fischbach,et al.  Microbial Transplantation With Human Gut Commensals Containing CutC Is Sufficient to Transmit Enhanced Platelet Reactivity and Thrombosis Potential , 2018, Circulation research.

[55]  Chunyan Wu,et al.  A metagenomic study of the gut microbiome in Behcet’s disease , 2018, Microbiome.

[56]  Min-jung Park,et al.  Attenuation of Rheumatoid Inflammation by Sodium Butyrate Through Reciprocal Targeting of HDAC2 in Osteoclasts and HDAC8 in T Cells , 2018, Front. Immunol..

[57]  H. Blottière,et al.  Butyrate produced by gut commensal bacteria activates TGF-beta1 expression through the transcription factor SP1 in human intestinal epithelial cells , 2018, Scientific Reports.

[58]  S. Hazen,et al.  Development of a gut microbe-targeted non-lethal therapeutic to inhibit thrombosis potential , 2018, Nature Medicine.

[59]  Mingming Zhao,et al.  Gut flora-dependent metabolite Trimethylamine-N-oxide accelerates endothelial cell senescence and vascular aging through oxidative stress. , 2018, Free radical biology & medicine.

[60]  T. Kim,et al.  Rediscover the clinical value of small intestinal bacterial overgrowth in patients with intestinal Behçet's disease , 2018, Journal of gastroenterology and hepatology.

[61]  M. Mohajeri,et al.  Probiotics and the Gut Immune System: Indirect Regulation , 2017, Probiotics and Antimicrobial Proteins.

[62]  C. Lebrilla,et al.  Microbiota-activated PPAR-γ signaling inhibits dysbiotic Enterobacteriaceae expansion , 2017, Science.

[63]  W. Liao,et al.  Influence of diet on the gut microbiome and implications for human health , 2017, Journal of Translational Medicine.

[64]  Ashwin N Ananthakrishnan,et al.  Gut Microbiome Function Predicts Response to Anti-integrin Biologic Therapy in Inflammatory Bowel Diseases. , 2017, Cell host & microbe.

[65]  Mingming Zhao,et al.  Trimethylamine N-oxide in atherogenesis: impairing endothelial self-repair capacity and enhancing monocyte adhesion , 2017, Bioscience reports.

[66]  S. Hazen,et al.  Microbiome, trimethylamine N-oxide, and cardiometabolic disease. , 2017, Translational research : the journal of laboratory and clinical medicine.

[67]  F. Cianchi,et al.  Cytotoxic Th1 and Th17 cells infiltrate the intestinal mucosa of Behcet patients and exhibit high levels of TNF-α in early phases of the disease , 2016, Medicine.

[68]  T. Ergun,et al.  Sequencing of 16S rRNA reveals a distinct salivary microbiome signature in Behçet's disease. , 2016, Clinical immunology.

[69]  J. Shimizu,et al.  Bifidobacteria Abundance-Featured Gut Microbiota Compositional Change in Patients with Behcet’s Disease , 2016, PloS one.

[70]  Renan Corrêa-Oliveira,et al.  Regulation of immune cell function by short-chain fatty acids , 2016, Clinical & translational immunology.

[71]  S. Hazen,et al.  Gut Microbial Metabolite TMAO Enhances Platelet Hyperreactivity and Thrombosis Risk , 2016, Cell.

[72]  S. Hazen,et al.  Trimethylamine N‐Oxide Promotes Vascular Inflammation Through Signaling of Mitogen‐Activated Protein Kinase and Nuclear Factor‐κB , 2016, Journal of the American Heart Association.

[73]  Michael A. Fischbach,et al.  A biosynthetic pathway for a prominent class of microbiota-derived bile acids , 2015, Nature chemical biology.

[74]  S. Rampelli,et al.  Behçet's syndrome patients exhibit specific microbiome signature. , 2015, Autoimmunity reviews.

[75]  L. Bergmeier,et al.  The oral mucosal and salivary microbial community of Behçet's syndrome and recurrent aphthous stomatitis , 2015, Journal of oral microbiology.

[76]  S. Kang,et al.  Short chain fatty acids induce both effector and regulatory T cells by suppression of histone deacetylases and regulation of the mTOR-S6K pathway , 2014, Mucosal Immunology.

[77]  S. Hazen,et al.  γ-Butyrobetaine is a proatherogenic intermediate in gut microbial metabolism of L-carnitine to TMAO. , 2014, Cell metabolism.

[78]  Y. Belkaid,et al.  Role of the Microbiota in Immunity and Inflammation , 2014, Cell.

[79]  M. Tomita,et al.  Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells , 2013, Nature.

[80]  J. Gordon,et al.  Olfactory receptor responding to gut microbiota-derived signals plays a role in renin secretion and blood pressure regulation , 2013, Proceedings of the National Academy of Sciences.

[81]  E. Balskus,et al.  Microbial conversion of choline to trimethylamine requires a glycyl radical enzyme , 2012, Proceedings of the National Academy of Sciences.

[82]  Katherine H. Huang,et al.  Structure, Function and Diversity of the Healthy Human Microbiome , 2012, Nature.

[83]  P. Tontonoz,et al.  Transcriptional integration of metabolism by the nuclear sterol-activated receptors LXR and FXR , 2012, Nature Reviews Molecular Cell Biology.

[84]  T. Karagiannis,et al.  Histone Deacetylase Inhibition and Dietary Short-Chain Fatty Acids , 2011, ISRN allergy.

[85]  Wei Sun,et al.  The microbiome and butyrate regulate energy metabolism and autophagy in the mammalian colon. , 2011, Cell metabolism.

[86]  Brian J. Bennett,et al.  Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease , 2011, Nature.

[87]  Stanley L Hazen,et al.  Plasma myeloperoxidase predicts incident cardiovascular risks in stable patients undergoing medical management for coronary artery disease. , 2011, Clinical chemistry.

[88]  B. M. Forman,et al.  Farnesoid X receptor antagonizes nuclear factor κB in hepatic inflammatory response , 2008, Hepatology.

[89]  R Balfour Sartor,et al.  Specific microbiota direct the differentiation of IL-17-producing T-helper cells in the mucosa of the small intestine. , 2008, Cell host & microbe.

[90]  T. Warner,et al.  Farnesoid X Receptor Ligands Inhibit Vascular Smooth Muscle Cell Inflammation and Migration , 2007, Arteriosclerosis, thrombosis, and vascular biology.

[91]  Stefan Offermanns,et al.  Activation of Platelet Function Through G Protein–Coupled Receptors Platelets As Immune Cells: Bridging Inflammation and Cardiovascular Disease In Vivo Thrombus Formation Platelet Adhesion Platelet Inhibitors and Thrombus Formation , 2006 .

[92]  Dae-Joong Kang,et al.  Bile salt biotransformations by human intestinal bacteria Published, JLR Papers in Press, November 18, 2005. , 2006, Journal of Lipid Research.

[93]  W. Hörl,et al.  Anti‐inflammatory effects of sodium butyrate on human monocytes: potent inhibition of IL‐12 and up‐regulation of IL‐10 production , 2000, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[94]  D. Williamson,et al.  Tissue and bacterial splitting of sulphasalazine. , 1983, Clinical science.