Fecal microbiota transplantation alleviates experimental colitis through the Toll-like receptor 4 signaling pathway

BACKGROUND Fecal microbiota transplantation (FMT) has shown promising therapeutic effects on mice with experimental colitis and patients with ulcerative colitis (UC). FMT modulates the Toll-like receptor 4 (TLR4) signaling pathway to treat some other diseases. However, it remains unknown whether this modulation is also involved in the treatment of UC. AIM To clarify the necessity of TLR4 signaling pathway in FMT on dextran sodium sulphate (DSS)-induced mice and explain the mechanism of FMT on UC, through association analysis of gut microbiota with colon transcriptome in mice. METHODS A mouse colitis model was constructed with wild-type (WT) and TLR4-knockout (KO) mice. Fecal microbiota was transplanted by gavage. Colon inflammation severity was measured by disease activity index (DAI) scoring and hematoxylin and eosin staining. Gut microbiota structure was analyzed through 16S ribosomal RNA sequencing. Gene expression in the mouse colon was obtained by transcriptome sequencing. RESULTS The KO (DSS + Water) and KO (DSS + FMT) groups displayed indistinguishable body weight loss, colon length, DAI score, and histology score, which showed that FMT could not inhibit the disease in KO mice. In mice treated with FMT, the relative abundance of Akkermansia decreased, and Lactobacillus became dominant. In particular, compared with those in WT mice, the scores of DAI and colon histology were clearly decreased in the KO-DSS group. Microbiota structure showed a significant difference between KO and WT mice. Akkermansia were the dominant genus in healthy KO mice. The ineffectiveness of FMT in KO mice was related to the decreased abundance of Akkermansia. Gene Ontology enrichment analysis showed that differentially expressed genes between each group were mainly involved in cytoplasmic translation and cellular response to DNA damage stimulus. The top nine genes correlating with Akkermansia included Aqp4, Clca4a, Dpm3, Fau, Mcrip1, Meis3, Nupr1 L, Pank3, and Rps13 (|R| > 0.9, P < 0.01). CONCLUSION FMT may ameliorate DSS-induced colitis by regulating the TLR4 signaling pathway. TLR4 modulates the composition of gut microbiota and the expression of related genes to ameliorate colitis and maintain the stability of the intestinal environment. Akkermansia bear great therapeutic potential for colitis.

[1]  S. Alshawwa,et al.  The immunomodulatory effects of probiotics and azithromycin in dextran sodium sulfate-induced ulcerative colitis in rats via TLR4-NF-κB and p38-MAPK pathway. , 2023, Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie.

[2]  Feng-jie Tian,et al.  Mitigation of maternal fecal microbiota transplantation on neurobehavioral deficits of offspring rats prenatally exposed to arsenic: Role of microbiota-gut-brain axis. , 2023, Journal of hazardous materials.

[3]  R. Niu,et al.  Intestinal microbiota regulates colonic inflammation in fluorosis mice by TLR/NF-κB pathway through short-chain fatty acids. , 2023, Food and chemical toxicology : an international journal published for the British Industrial Biological Research Association.

[4]  H. Wen,et al.  Structurally dynamic self-healable hydrogel cooperatively inhibits intestinal inflammation and promotes mucosal repair for enhanced ulcerative colitis treatment. , 2023, Biomaterials.

[5]  H. Yang,et al.  Lactobacillus rhamnosus and L. plantarum combination treatment ameliorated colitis symptoms in a mouse model by altering intestinal microbial composition and suppressing inflammatory response. , 2023, Molecular nutrition & food research.

[6]  Shanshan Li,et al.  Blueberry extract alleviated lipopolysaccharide-induced inflammation responses in mice through activating the FXR/TGR5 signaling pathway and regulating gut microbiota. , 2023, Journal of the science of food and agriculture.

[7]  Lei Tang,et al.  Akkermansia muciniphila attenuates LPS-induced acute kidney injury by inhibiting TLR4/NF-κB pathway. , 2022, FEMS microbiology letters.

[8]  Shangyong Li,et al.  Preventive effect of pectic oligosaccharides on acute colitis model mice: modulating epithelial barrier, gut microbiota and Treg/Th17 balance. , 2022, Food & function.

[9]  X. Pang,et al.  Characterization of the Fungal Community in Fritillariae Cirrhosae Bulbus through DNA Metabarcoding , 2022, Journal of fungi.

[10]  Wei Wang,et al.  TLR4 regulates RORγt+ regulatory T-cell responses and susceptibility to colon inflammation through interaction with Akkermansia muciniphila , 2022, Microbiome.

[11]  Xiao-Zhong Yang,et al.  Cepharanthine ameliorates dextran sulphate sodium‐induced colitis through modulating gut microbiota , 2022, Microbial biotechnology.

[12]  X. Chen,et al.  Fecal Microbiota Transplantation Ameliorates Active Ulcerative Colitis by Downregulating Pro-inflammatory Cytokines in Mucosa and Serum , 2022, Frontiers in Microbiology.

[13]  K. Xu,et al.  Atractyloside-A ameliorates spleen deficiency diarrhea by interfering with TLR4/MyD88/NF-κB signaling activation and regulating intestinal flora homeostasis. , 2022, International immunopharmacology.

[14]  Dan Zhang,et al.  Fecal microbiota transplantation protects rotenone-induced Parkinson’s disease mice via suppressing inflammation mediated by the lipopolysaccharide-TLR4 signaling pathway through the microbiota-gut-brain axis , 2021, Microbiome.

[15]  G. Putzel,et al.  Transferable IgA-coated Odoribacter splanchnicus in Responders to Fecal Microbiota Transplantation for Ulcerative Colitis Limits Colonic Inflammation. , 2021, Gastroenterology.

[16]  Z. Xu,et al.  Gut microbiota from green tea polyphenol-dosed mice improves intestinal epithelial homeostasis and ameliorates experimental colitis , 2021, Microbiome.

[17]  Jie Guo,et al.  TLR4 signaling in the development of colitis-associated cancer and its possible interplay with microRNA-155 , 2021, Cell communication and signaling : CCS.

[18]  Huan Gao,et al.  Fecal Microbiota Transplantation Exerts a Protective Role in MPTP-Induced Parkinson’s Disease via the TLR4/PI3K/AKT/NF-κB Pathway Stimulated by α-Synuclein , 2021, Neurochemical Research.

[19]  Ting Zhang,et al.  The potential of Akkermansia muciniphila in inflammatory bowel disease , 2021, Applied Microbiology and Biotechnology.

[20]  Xiao-Zhong Yang,et al.  Fecal microbiota transplantation ameliorates experimental colitis via gut microbiota and T-cell modulation , 2021, World journal of gastroenterology.

[21]  Yi-nan Zhao,et al.  Shaoyao-Gancao Decoction Ameliorates the Inflammation State in Polycystic Ovary Syndrome Rats via Remodeling Gut Microbiota and Suppressing the TLR4/NF-κB Pathway , 2021, Frontiers in Pharmacology.

[22]  M. Kalady,et al.  Ketogenic diet alleviates colitis by reduction of colonic group 3 innate lymphoid cells through altering gut microbiome , 2021, Signal Transduction and Targeted Therapy.

[23]  F. Du,et al.  Metagenome Analysis of Intestinal Bacteria in Healthy People, Patients With Inflammatory Bowel Disease and Colorectal Cancer , 2021, Frontiers in Cellular and Infection Microbiology.

[24]  Baoming Tian,et al.  Lycium ruthenicum anthocyanins attenuate high-fat diet-induced colonic barrier dysfunction and inflammation in mice by modulating the gut microbiota. , 2021, Molecular nutrition & food research.

[25]  C. Prestidge,et al.  Toll-like receptor 4 (TLR4) antagonists as potential therapeutics for intestinal inflammation , 2021, Indian Journal of Gastroenterology.

[26]  Wei-Yu Lu,et al.  Relationship Between Pregnancy and Acute Disseminated Encephalomyelitis: A Single-Case Study , 2021, Frontiers in Immunology.

[27]  Jia-feng Tang,et al.  Effect of gut microbiota on LPS-induced acute lung injury by regulating the TLR4/NF-kB signaling pathway. , 2020, International immunopharmacology.

[28]  S. Mehandru,et al.  The intestinal barrier, an arbitrator turned provocateur in IBD , 2020, Nature Reviews Gastroenterology & Hepatology.

[29]  A. Nierenberg,et al.  Efficacy and safety of fecal microbiota transplantation for the treatment of diseases other than Clostridium difficile infection: a systematic review and meta-analysis , 2020, Gut microbes.

[30]  M. Peitsch,et al.  Inflammatory Bowel Disease–Associated Changes in the Gut: Focus on Kazan Patients , 2020, Inflammatory bowel diseases.

[31]  J. Duan,et al.  Lizhong decoction ameliorates ulcerative colitis in mice via modulating gut microbiota and its metabolites , 2020, Applied Microbiology and Biotechnology.

[32]  Liyong Luo,et al.  The Prebiotic Properties of Green and Dark Tea Contribute to The Protective Effects in Chemical-Induced Colitis in Mice: A Fecal Microbiota Transplantation Study. , 2020, Journal of agricultural and food chemistry.

[33]  Xiaofeng Jiang,et al.  Fecal Microbiota Transplantation (FMT) Alleviates Experimental Colitis in Mice by Gut Microbiota Regulation , 2020, Journal of microbiology and biotechnology.

[34]  Lijuan Wang,et al.  A purified membrane protein from Akkermansia muciniphila or the pasteurised bacterium blunts colitis associated tumourigenesis by modulation of CD8+ T cells in mice , 2020, Gut.

[35]  Ziren Su,et al.  Oxyberberine, a novel gut microbiota-mediated metabolite of berberine, possesses superior anti-colitis effect: impact on intestinal epithelial barrier, gut microbiota profile and TLR4-MyD88-NF-κB pathway. , 2019, Pharmacological research.

[36]  M. Surette,et al.  Differences in Gut Microbiota in Patients With vs Without Inflammatory Bowel Diseases: a Systematic Review. , 2019, Gastroenterology.

[37]  Yongmin Yan,et al.  Mesenchymal stem cell–gut microbiota interaction in the repair of inflammatory bowel disease: an enhanced therapeutic effect , 2019, Clinical and Translational Medicine.

[38]  Xiao-shuang Liu,et al.  Critical Role of Toll-Like Receptor 4 (TLR4) in Dextran Sulfate Sodium (DSS)-Induced Intestinal Injury and Repair. , 2019, Toxicology letters.

[39]  Lanjuan Li,et al.  Administration of Akkermansia muciniphila Ameliorates Dextran Sulfate Sodium-Induced Ulcerative Colitis in Mice , 2019, Front. Microbiol..

[40]  H. Vlamakis,et al.  Microbial genes and pathways in inflammatory bowel disease , 2019, Nature Reviews Microbiology.

[41]  Ying Xie,et al.  Palmatine ameliorated murine colitis by suppressing tryptophan metabolism and regulating gut microbiota , 2018, Pharmacological research.

[42]  Xi Chen,et al.  PathwaySplice: an R package for unbiased pathway analysis of alternative splicing in RNA-Seq data , 2018, Bioinform..

[43]  E. Hsiao,et al.  The Gut Microbiota Mediates the Anti-Seizure Effects of the Ketogenic Diet , 2018, Cell.

[44]  Bradley W. Bolling,et al.  A common antimicrobial additive increases colonic inflammation and colitis-associated colon tumorigenesis in mice , 2018, Science Translational Medicine.

[45]  Xun Ma,et al.  Inhibition of HMGB1 reduces rat spinal cord astrocytic swelling and AQP4 expression after oxygen-glucose deprivation and reoxygenation via TLR4 and NF-κB signaling in an IL-6-dependent manner , 2017, Journal of Neuroinflammation.

[46]  L. Albenberg,et al.  Gut microbiota and IBD: causation or correlation? , 2017, Nature Reviews Gastroenterology &Hepatology.

[47]  Hugh Thomas IBD: FMT induces clinical remission in ulcerative colitis , 2017, Nature Reviews Gastroenterology &Hepatology.

[48]  M. Eisenhut,et al.  IBD immunopathogenesis: A comprehensive review of inflammatory molecules. , 2017, Autoimmunity reviews.

[49]  J. Versalovic,et al.  Composition and function of the pediatric colonic mucosal microbiome in untreated patients with ulcerative colitis , 2016, Gut microbes.

[50]  Xue-qun Chen,et al.  Systemic pro-inflammatory response facilitates the development of cerebral edema during short hypoxia , 2016, Journal of Neuroinflammation.

[51]  M. Surette,et al.  Fecal Microbiota Transplantation Induces Remission in Patients With Active Ulcerative Colitis in a Randomized Controlled Trial. , 2015, Gastroenterology.

[52]  Gongping Xu,et al.  Curcumin improves the recovery of motor function and reduces spinal cord edema in a rat acute spinal cord injury model by inhibiting the JAK/STAT signaling pathway. , 2014, Acta histochemica.

[53]  K. Yasukawa,et al.  Involvement of nitric oxide with activation of Toll-like receptor 4 signaling in mice with dextran sodium sulfate-induced colitis. , 2014, Free radical biology & medicine.

[54]  M. Papadopoulos,et al.  Aquaporins: important but elusive drug targets , 2014, Nature Reviews Drug Discovery.

[55]  Robert C. Edgar,et al.  UPARSE: highly accurate OTU sequences from microbial amplicon reads , 2013, Nature Methods.

[56]  L. Bilston,et al.  Aquaporin-4 expression in post-traumatic syringomyelia. , 2013, Journal of neurotrauma.

[57]  B. Kuster,et al.  Lactocepin secreted by Lactobacillus exerts anti-inflammatory effects by selectively degrading proinflammatory chemokines. , 2012, Cell host & microbe.

[58]  M. Papadopoulos,et al.  Aquaporin-4 in brain and spinal cord oedema , 2010, Neuroscience.

[59]  Matthew D. Young,et al.  Gene ontology analysis for RNA-seq: accounting for selection bias , 2010, Genome Biology.

[60]  Yoshihiro Yamanishi,et al.  KEGG for linking genomes to life and the environment , 2007, Nucleic Acids Res..

[61]  R. Ley,et al.  Ecological and Evolutionary Forces Shaping Microbial Diversity in the Human Intestine , 2006, Cell.

[62]  Tao Cai,et al.  Automated genome annotation and pathway identification using the KEGG Orthology (KO) as a controlled vocabulary , 2005, Bioinform..

[63]  Erko Stackebrandt,et al.  Taxonomic Note: A Place for DNA-DNA Reassociation and 16S rRNA Sequence Analysis in the Present Species Definition in Bacteriology , 1994 .

[64]  Yan Tan 谭 琰,et al.  Expression and implication of toll-like receptors TLR2, TLR4 and TLR9 in colonic mucosa of patients with ulcerative colitis , 2014, Journal of Huazhong University of Science and Technology [Medical Sciences].

[65]  Jyoti Das,et al.  Inflammatory bowel disease requires the interplay between innate and adaptive immune signals , 2006, Cell Research.