Levofloxacin induces differential effects in the transcriptome between the gut, peripheral and axial joints in the Spondyloarthritis DBA/1 mice: Improvement of intestinal dysbiosis and the overall inflammatory process

To analyze the effect of levofloxacin-induced intestinal microbiota modifications on intestinal, joint, and systemic inflammation in the DBA/1 mice with spontaneous arthritis. The study included two groups of mice, one of which received levofloxacin. The composition and structure of the microbiota were determined in the mice’s stool using 16S rRNA sequencing; the differential taxa and metabolic pathway between mice treated with levofloxacin and control mice were also defied. The effect of levofloxacin was evaluated in the intestines, hind paws, and spines of mice through DNA microarray transcriptome and histopathological analyses; systemic inflammation was measured by flow cytometry. Levofloxacin decreased the pro-inflammatory bacteria, including Prevotellaceae, Odoribacter, and Blautia, and increased the anti-inflammatory Muribaculaceae in mice’s stool. Histological analysis confirmed the intestinal inflammation in control mice, while in levofloxacin-treated mice, inflammation was reduced; in the hind paws and spines, levofloxacin also decreased the inflammation. Microarray showed the downregulation of genes and signaling pathways relevant in spondyloarthritis, including several cytokines and chemokines. Levofloxacin-treated mice showed differential transcriptomic profiles between peripheral and axial joints and intestines. Levofloxacin decreased the expression of TNF-α, IL-23a, and JAK3 in the three tissues, but IL-17 behaved differently in the intestine and the joints. Serum TNF-α was also reduced in levofloxacin-treated mice. Our results suggest that the microbiota modification aimed at reducing pro-inflammatory and increasing anti-inflammatory bacteria could potentially be a coadjuvant in treating inflammatory arthropathies.

[1]  Lei Wang,et al.  A polysaccharide from Rosa roxburghii Tratt fruit attenuates high-fat diet-induced intestinal barrier dysfunction and inflammation in mice by modulating the gut microbiota. , 2021, Food & function.

[2]  Peizhan Chen,et al.  Ursodeoxycholic Acid Treatment Restores Gut Microbiota and Alleviates Liver Inflammation in Non-Alcoholic Steatohepatitic Mouse Model , 2021, Frontiers in Pharmacology.

[3]  Qiqi Liu,et al.  Effect of Extracellular Vesicles Derived From Lactobacillus plantarum Q7 on Gut Microbiota and Ulcerative Colitis in Mice , 2021, Frontiers in Immunology.

[4]  Y. Tuo,et al.  Exopolysaccharide Produced by Lactiplantibacillus plantarum-12 Alleviates Intestinal Inflammation and Colon Cancer Symptoms by Modulating the Gut Microbiome and Metabolites of C57BL/6 Mice Treated by Azoxymethane/Dextran Sulfate Sodium Salt , 2021, Foods.

[5]  B. Xiao,et al.  A High-Tryptophan Diet Reduces Seizure-Induced Respiratory Arrest and Alters the Gut Microbiota in DBA/1 Mice , 2021, Frontiers in Neurology.

[6]  B. Ren,et al.  Novel Sesquiterpene Glycoside from Loquat Leaf Alleviates Type 2 Diabetes Mellitus Combined with Nonalcoholic Fatty Liver Disease by Improving Insulin Resistance, Oxidative Stress, Inflammation, and Gut Microbiota Composition. , 2021, Journal of agricultural and food chemistry.

[7]  Ming Yang,et al.  Pulsatilla chinensis Saponins Ameliorate Inflammation and DSS-Induced Ulcerative Colitis in Rats by Regulating the Composition and Diversity of Intestinal Flora , 2021, Frontiers in Cellular and Infection Microbiology.

[8]  R. Landewé,et al.  No efficacy of anti-IL-23 therapy for axial spondyloarthritis in randomised controlled trials but in post-hoc analyses of psoriatic arthritis-related ‘physician-reported spondylitis’? , 2021, Annals of the Rheumatic Diseases.

[9]  Courtney M. Mazur,et al.  Mechanosensitive miR‐100 coordinates TGFβ and Wnt signaling in osteocytes during fluid shear stress , 2021, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[10]  E. Martens,et al.  Leveraging diet to engineer the gut microbiome , 2021, Nature Reviews Gastroenterology & Hepatology.

[11]  G. Barbara,et al.  Inflammatory and Microbiota-Related Regulation of the Intestinal Epithelial Barrier , 2021, Frontiers in Nutrition.

[12]  A. Silman,et al.  ‘All disease begins in the gut’—the role of the intestinal microbiome in ankylosing spondylitis , 2021, Rheumatology advances in practice.

[13]  A. Jabłońska,et al.  The Role of Fecal Microbiota Transplantation in the Treatment of Inflammatory Bowel Disease , 2021, Journal of clinical medicine.

[14]  P. Sarzi-Puttini,et al.  Intestinal microbiota changes induced by TNF-inhibitors in IBD-related spondyloarthritis , 2021, RMD Open.

[15]  P. Kashyap,et al.  The promise of the gut microbiome as part of individualized treatment strategies , 2021, Nature Reviews Gastroenterology & Hepatology.

[16]  B. Mavčič,et al.  The role of molecular diagnostics in aneurysmal and simple bone cysts – a prospective analysis of 19 lesions , 2021, Virchows Archiv.

[17]  Xiaojuan Yang,et al.  Role of Fzd6 in Regulating the Osteogenic Differentiation of Adipose-derived Stem Cells in Osteoporotic Mice , 2021, Stem Cell Reviews and Reports.

[18]  S. Millis,et al.  Multiscale-omic assessment of EWSR1-NFATc2 fusion positive sarcomas identifies the mTOR pathway as a potential therapeutic target , 2021, npj Precision Oncology.

[19]  P. Boor,et al.  Aberrant Gut Microbiome Contributes to Intestinal Oxidative Stress, Barrier Dysfunction, Inflammation and Systemic Autoimmune Responses in MRL/lpr Mice , 2021, Frontiers in Immunology.

[20]  Magdalena Ciążyńska,et al.  Alterations of the Skin and Gut Microbiome in Psoriasis and Psoriatic Arthritis , 2021, International journal of molecular sciences.

[21]  Ibis M Agosto-Marlin,et al.  New Studies of Pathogenesis of Rheumatoid Arthritis with Collagen-Induced and Collagen Antibody-Induced Arthritis Models: New Insight Involving Bacteria Flora , 2021, Autoimmune diseases.

[22]  C. Kavadichanda,et al.  Spondyloarthritis and the Human Leukocyte Antigen (HLA)-B*27 Connection , 2021, Frontiers in Immunology.

[23]  Xiaobin Pang,et al.  Ginger Alleviates DSS-Induced Ulcerative Colitis Severity by Improving the Diversity and Function of Gut Microbiota , 2021, Frontiers in Pharmacology.

[24]  I. Guénal,et al.  Lessons on SpA pathogenesis from animal models , 2021, Seminars in immunopathology.

[25]  P. Hugenholtz,et al.  Streptococcus species enriched in the oral cavity of patients with RA are a source of peptidoglycan-polysaccharide polymers that can induce arthritis in mice , 2021, Annals of the Rheumatic Diseases.

[26]  Nadezhda T. Doncheva,et al.  The STRING database in 2021: customizable protein–protein networks, and functional characterization of user-uploaded gene/measurement sets , 2020, Nucleic Acids Res..

[27]  Dongjie Zhang,et al.  Potential effects of mung bean protein and a mung bean protein-polyphenol complex on oxidative stress levels and intestinal microflora in aging mice , 2021, Food & Function.

[28]  A. Iafrate,et al.  Identification of EWSR1–NFATC2 fusion in simple bone cysts , 2020, Histopathology.

[29]  Xiaoying Lin,et al.  Combined effects of MSU crystals injection and high fat-diet feeding on the establishment of a gout model in C57BL/6 mice , 2020, Advances in Rheumatology.

[30]  L. Tam,et al.  Gut Microbiome and Its Interaction with Immune System in Spondyloarthritis , 2020, Microorganisms.

[31]  P. Bowness,et al.  New developments in our understanding of ankylosing spondylitis pathogenesis , 2020, Immunology.

[32]  Byron J. Smith,et al.  Muribaculaceae Genomes Assembled from Metagenomes Suggest Genetic Drivers of Differential Response to Acarbose Treatment in Mice , 2020, bioRxiv.

[33]  Gavin M Douglas,et al.  PICRUSt2 for prediction of metagenome functions , 2020, Nature Biotechnology.

[34]  Chaofei Yang,et al.  MACF1 promotes preosteoblast migration by mediating focal adhesion turnover through EB1 , 2020, Biology Open.

[35]  Q. Ou,et al.  Alteration of the gut microbiota in tumor necrosis factor‐α antagonist‐treated collagen‐induced arthritis mice , 2020, International journal of rheumatic diseases.

[36]  Jianguo Xia,et al.  Using MicrobiomeAnalyst for comprehensive statistical, functional, and meta-analysis of microbiome data , 2020, Nature Protocols.

[37]  Cui-li Ma,et al.  Dynamic Variations in Gut Microbiota in Ankylosing Spondylitis Patients Treated with Anti-TNF-α for Six Months. , 2020, Annals of clinical and laboratory science.

[38]  F. Ciccia,et al.  Gut dysbiosis in Spondyloarthritis: Cause or effect? , 2019, Best practice & research. Clinical rheumatology.

[39]  M. Breban,et al.  The microbiome in spondyloarthritis. , 2019, Best practice & research. Clinical rheumatology.

[40]  Zhuye Jie,et al.  Metagenomic profiling of the pro-inflammatory gut microbiota in ankylosing spondylitis. , 2019, Journal of autoimmunity.

[41]  L. Öhman,et al.  A distinct gut microbiota composition in patients with ankylosing spondylitis is associated with increased levels of fecal calprotectin , 2019, Arthritis Research & Therapy.

[42]  M. J. Kim,et al.  Correlation of Dickkopf-1 with Inflammation in Crohn Disease , 2019, Indian Pediatrics.

[43]  M. Asquith,et al.  Novel Inter‐omic Analysis Reveals Relationships Between Diverse Gut Microbiota and Host Immune Dysregulation in HLA–B27–Induced Experimental Spondyloarthritis , 2019, Arthritis & rheumatology.

[44]  A. Uitterlinden,et al.  Intestinal microbiome composition and its relation to joint pain and inflammation , 2019, Nature Communications.

[45]  Z. Pei,et al.  Characteristics of Gut Microbiota in Patients With Rheumatoid Arthritis in Shanghai, China , 2019, Front. Cell. Infect. Microbiol..

[46]  F. Pan,et al.  Fecal microbiota in patients with ankylosing spondylitis: Correlation with dietary factors and disease activity. , 2019, Clinica chimica acta; international journal of clinical chemistry.

[47]  Francesco Asnicar,et al.  Reproducible, interactive, scalable and extensible microbiome data science using QIIME 2 , 2019, Nature Biotechnology.

[48]  Byron J. Smith,et al.  Changes in the gut microbiome and fermentation products concurrent with enhanced longevity in acarbose-treated mice , 2019, BMC Microbiology.

[49]  N. Phillips,et al.  Familial Adenomatous Polyposis Syndrome: An Update and Review of Extraintestinal Manifestations. , 2019, Archives of pathology & laboratory medicine.

[50]  S. Feldman,et al.  Mechanisms of microbial pathogenesis and the role of the skin microbiome in psoriasis: A review. , 2019, Clinics in dermatology.

[51]  F. Guilak,et al.  Physiologic and pathologic effects of dietary free fatty acids on cells of the joint , 2019, Annals of the New York Academy of Sciences.

[52]  B. Stecher,et al.  Sequence and cultivation study of Muribaculaceae reveals novel species, host preference, and functional potential of this yet undescribed family , 2019, Microbiome.

[53]  Chris C. Tang,et al.  Compositional changes to the ileal microbiome precede the onset of spontaneous ileitis in SHIP deficient mice , 2019, Gut microbes.

[54]  D. Heijde,et al.  Three Multicenter, Randomized, Double‐Blind, Placebo‐Controlled Studies Evaluating the Efficacy and Safety of Ustekinumab in Axial Spondyloarthritis , 2018, Arthritis & rheumatology.

[55]  R. Marrie,et al.  A comparative study of the gut microbiota in immune-mediated inflammatory diseases—does a common dysbiosis exist? , 2018, Microbiome.

[56]  Man Xiao,et al.  Protective effects of Paederia scandens extract on rheumatoid arthritis mouse model by modulating gut microbiota. , 2018, Journal of ethnopharmacology.

[57]  Kyongbum Lee,et al.  Parabacteroides distasonis attenuates toll‐like receptor 4 signaling and Akt activation and blocks colon tumor formation in high‐fat diet‐fed azoxymethane‐treated mice , 2018, International journal of cancer.

[58]  D. Frank,et al.  Modulation of Inflammatory Arthritis in Mice by Gut Microbiota Through Mucosal Inflammation and Autoantibody Generation , 2018, Arthritis & rheumatology.

[59]  M. Danila,et al.  Age and fecal microbial strain-specific differences in patients with spondyloarthritis , 2018, Arthritis Research & Therapy.

[60]  Gavin M Douglas,et al.  Processing a 16S rRNA Sequencing Dataset with the Microbiome Helper Workflow. , 2018, Methods in molecular biology.

[61]  S. Abramson,et al.  Alteration of the intestinal microbiome characterizes preclinical inflammatory arthritis in mice and its modulation attenuates established arthritis , 2017, Scientific Reports.

[62]  Damian Szklarczyk,et al.  The STRING database in 2017: quality-controlled protein–protein association networks, made broadly accessible , 2016, Nucleic Acids Res..

[63]  C. Donati,et al.  Alteration of Fecal Microbiota Profiles in Juvenile Idiopathic Arthritis. Associations with HLA-B27 Allele and Disease Status , 2016, Frontiers in Microbiology.

[64]  B. Zeng,et al.  Role of the Gut Microbiome in Modulating Arthritis Progression in Mice , 2016, Scientific Reports.

[65]  S. González-Chávez,et al.  Molecular mechanisms of bone formation in spondyloarthritis. , 2016, Joint, bone, spine : revue du rhumatisme.

[66]  J. Fonseca,et al.  Review: Animal Models as a Tool to Dissect Pivotal Pathways Driving Spondyloarthritis , 2015, Arthritis & rheumatology.

[67]  S. González-Chávez,et al.  The Danger Model Approach to the Pathogenesis of the Rheumatic Diseases , 2015, Journal of immunology research.

[68]  Mhairi Marshall,et al.  Brief Report: Intestinal Dysbiosis in Ankylosing Spondylitis , 2014, Arthritis & rheumatology.

[69]  Xiangqin Cui,et al.  Altered microbiota associated with abnormal humoral immune responses to commensal organisms in enthesitis-related arthritis , 2014, Arthritis Research & Therapy.

[70]  Aaron Y. Lee,et al.  HLA-B27 and Human β2-Microglobulin Affect the Gut Microbiota of Transgenic Rats , 2014, PloS one.

[71]  C. Loddenkemper,et al.  A guide to histomorphological evaluation of intestinal inflammation in mouse models. , 2014, International journal of clinical and experimental pathology.

[72]  Gary D. Bader,et al.  Enrichment Map – a Cytoscape app to visualize and explore OMICs pathway enrichment results , 2014, F1000Research.

[73]  M. Krausová,et al.  Wnt signaling in adult intestinal stem cells and cancer. , 2014, Cellular signalling.

[74]  S. González-Chávez,et al.  Assessment of different decalcifying protocols on Osteopontin and Osteocalcin immunostaining in whole bone specimens of arthritis rat model by confocal immunofluorescence. , 2013, International journal of clinical and experimental pathology.

[75]  R. Lories,et al.  Spontaneous arthritis and ankylosis in male DBA/1 mice: further evidence for a role of behavioral factors in “stress-induced arthritis” , 2012, Biological Procedures Online.

[76]  Marc Vandemeulebroecke,et al.  Secukinumab, a human anti-IL-17A monoclonal antibody, for moderate to severe Crohn's disease: unexpected results of a randomised, double-blind placebo-controlled trial , 2012, Gut.

[77]  C. Huttenhower,et al.  Metagenomic biomarker discovery and explanation , 2011, Genome Biology.

[78]  T. Hudcovic,et al.  Oral administration of Parabacteroides distasonis antigens attenuates experimental murine colitis through modulation of immunity and microbiota composition , 2011, Clinical and experimental immunology.

[79]  C. Cheadle,et al.  Alternate protein kinase A activity identifies a unique population of stromal cells in adult bone , 2010, Proceedings of the National Academy of Sciences.

[80]  R. Baldassano,et al.  Extraintestinal manifestations of inflammatory bowel disease. , 2005, Minerva gastroenterologica e dietologica.

[81]  F. Luyten,et al.  Ankylosing enthesitis, dactylitis, and onychoperiostitis in male DBA/1 mice: a model of psoriatic arthritis , 2004, Annals of the rheumatic diseases.

[82]  P. Shannon,et al.  Cytoscape: a software environment for integrated models of biomolecular interaction networks. , 2003, Genome research.

[83]  Gary D. Bader,et al.  An automated method for finding molecular complexes in large protein interaction networks , 2003, BMC Bioinformatics.

[84]  K. Wilson,et al.  Differential Induction of Colitis and Gastritis in HLA-B27 Transgenic Rats Selectively Colonized with Bacteroides vulgatus or Escherichia coli , 1999, Infection and Immunity.

[85]  R. Hammer,et al.  Normal luminal bacteria, especially Bacteroides species, mediate chronic colitis, gastritis, and arthritis in HLA-B27/human beta2 microglobulin transgenic rats. , 1996, The Journal of clinical investigation.

[86]  L. Klareskog,et al.  Characterization of a spontaneously occurring arthritis in male DBA/1 mice. , 1992, Arthritis and rheumatism.

[87]  Foutch Pg,et al.  Extraintestinal manifestations of inflammatory bowel disease. , 1985, Arizona medicine.