Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
2.
Nat Commun ; 15(1): 7702, 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39231967

ABSTRACT

The human gut virome, which is mainly composed of bacteriophages, also includes viruses infecting archaea, yet their role remains poorly understood due to lack of isolates. Here, we characterize a temperate archaeal virus (MSTV1) infecting Methanobrevibacter smithii, the dominant methanogenic archaeon of the human gut. The MSTV1 genome is integrated in the host chromosome as a provirus which is sporadically induced, resulting in virion release. Using cryo-electron tomography, we capture several intracellular virion assembly intermediates and confirm that only a small fraction of the host population actively produces virions in vitro. Similar low frequency of induction is observed in a mouse colonization model, using mice harboring a stable consortium of 12 bacterial species (OMM12). Transcriptomic analysis suggests a regulatory lysogeny-lysis switch involving an interplay between viral proteins to maintain virus-host equilibrium, ensuring host survival and viral persistence. Thus, our study sheds light on archaeal virus-host interactions and highlights similarities with bacteriophages in establishing stable coexistence with their hosts in the gut.


Subject(s)
Archaeal Viruses , Gastrointestinal Microbiome , Methanobrevibacter , Animals , Humans , Methanobrevibacter/genetics , Methanobrevibacter/metabolism , Mice , Archaeal Viruses/genetics , Archaeal Viruses/physiology , Archaeal Viruses/ultrastructure , Genome, Viral/genetics , Virion/ultrastructure , Lysogeny , Female
3.
Gut Microbes ; 16(1): 2295384, 2024.
Article in English | MEDLINE | ID: mdl-38126163

ABSTRACT

The anaerobic bacterium Fusobacterium nucleatum is significantly associated with human colorectal cancer (CRC) and is considered a significant contributor to the disease. The mechanisms underlying the promotion of intestinal tumor formation by F. nucleatum have only been partially uncovered. Here, we showed that F. nucleatum releases a metabolite into the microenvironment that strongly activates NF-κB in intestinal epithelial cells via the ALPK1/TIFA/TRAF6 pathway. Furthermore, we showed that the released molecule had the biological characteristics of ADP-heptose. We observed that F. nucleatum induction of this pathway increased the expression of the inflammatory cytokine IL-8 and two anti-apoptotic genes known to be implicated in CRC, BIRC3 and TNFAIP3. Finally, it promoted the survival of CRC cells and reduced 5-fluorouracil chemosensitivity in vitro. Taken together, our results emphasize the importance of the ALPK1/TIFA pathway in Fusobacterium induced-CRC pathogenesis, and identify the role of ADP-H in this process.


Subject(s)
Colorectal Neoplasms , Gastrointestinal Microbiome , Humans , Fusobacterium nucleatum/metabolism , Base Composition , Phylogeny , RNA, Ribosomal, 16S , Sequence Analysis, DNA , Colorectal Neoplasms/pathology , Heptoses/metabolism , Tumor Microenvironment
4.
Microbiome ; 11(1): 46, 2023 03 09.
Article in English | MEDLINE | ID: mdl-36894986

ABSTRACT

BACKGROUND: Infections with SARS-CoV-2 have a pronounced impact on the gastrointestinal tract and its resident microbiome. Clear differences between severe cases of infection and healthy individuals have been reported, including the loss of commensal taxa. We aimed to understand if microbiome alterations including functional shifts are unique to severe cases or a common effect of COVID-19. We used high-resolution systematic multi-omic analyses to profile the gut microbiome in asymptomatic-to-moderate COVID-19 individuals compared to a control group. RESULTS: We found a striking increase in the overall abundance and expression of both virulence factors and antimicrobial resistance genes in COVID-19. Importantly, these genes are encoded and expressed by commensal taxa from families such as Acidaminococcaceae and Erysipelatoclostridiaceae, which we found to be enriched in COVID-19-positive individuals. We also found an enrichment in the expression of a betaherpesvirus and rotavirus C genes in COVID-19-positive individuals compared to healthy controls. CONCLUSIONS: Our analyses identified an altered and increased infective competence of the gut microbiome in COVID-19 patients. Video Abstract.


Subject(s)
COVID-19 , Gastrointestinal Microbiome , Microbiota , Humans , Gastrointestinal Microbiome/genetics , SARS-CoV-2/genetics , Multiomics
5.
Cell Host Microbe ; 30(9): 1201-1206, 2022 09 14.
Article in English | MEDLINE | ID: mdl-36108612

ABSTRACT

The human gut microbiome produces a functional complex of biomolecules, including nucleic acids, (poly)peptides, structural molecules, and metabolites. This impacts human physiology in multiple ways, especially by triggering inflammatory pathways in disease. At present, much remains to be learned about the identity of key effectors and their causal roles.


Subject(s)
Gastrointestinal Microbiome , Microbiota , Nucleic Acids , Humans , Microbiota/physiology
6.
Cell Host Microbe ; 30(9): 1340-1340.e1, 2022 09 14.
Article in English | MEDLINE | ID: mdl-36108614

ABSTRACT

The human gut microbiome is intricately connected to health and disease. Microbiome-derived molecules are implicated in many chronic diseases involving inflammation. Herein, we summarize the diverse complex of such immunogenic molecules, including nucleic acids, (poly)peptides, structural molecules, and metabolites. The interactions between this "expobiome" and human immune pathways are specifically illustrated in the context of chronic diseases. To view this SnapShot, open or download the PDF.


Subject(s)
Gastrointestinal Microbiome , Microbiota , Humans , Inflammation
7.
Gut Microbes ; 14(1): 2110639, 2022.
Article in English | MEDLINE | ID: mdl-36036242

ABSTRACT

The commensal bacteria that make up the gut microbiota impact the health of their host on multiple levels. In particular, the interactions taking place between the microbe-associated molecule patterns (MAMPs) and pattern recognition receptors (PRRs), expressed by intestinal epithelial cells (IECs), are crucial for maintaining intestinal homeostasis. While numerous studies showed that TLRs and NLRs are involved in the control of gut homeostasis by commensal bacteria, the role of additional innate immune receptors remains unclear. Here, we seek for novel MAMP-PRR interactions involved in the beneficial effect of the commensal bacterium Akkermansia muciniphila on intestinal homeostasis. We show that A. muciniphila strongly activates NF-κB in IECs by releasing one or more potent activating metabolites into the microenvironment. By using drugs, chemical and gene-editing tools, we found that the released metabolite(s) enter(s) epithelial cells and activate(s) NF-κB via an ALPK1, TIFA and TRAF6-dependent pathway. Furthermore, we show that the released molecule has the biological characteristics of the ALPK1 ligand ADP-heptose. Finally, we show that A. muciniphila induces the expression of the MUC2, BIRC3 and TNFAIP3 genes involved in the maintenance of the intestinal barrier function and that this process is dependent on TIFA. Altogether, our data strongly suggest that the commensal A. muciniphila promotes intestinal homeostasis by activating the ALPK1/TIFA/TRAF6 axis, an innate immune pathway exclusively described so far in the context of Gram-negative bacterial infections.


Subject(s)
Gastrointestinal Microbiome , NF-kappa B , Adenosine Diphosphate , Akkermansia , Heptoses , Immunity, Innate , TNF Receptor-Associated Factor 6 , Verrucomicrobia
8.
Nat Metab ; 4(4): 458-475, 2022 04.
Article in English | MEDLINE | ID: mdl-35437333

ABSTRACT

The gut microbiome is a key player in the immunomodulatory and protumorigenic microenvironment during colorectal cancer (CRC), as different gut-derived bacteria can induce tumour growth. However, the crosstalk between the gut microbiome and the host in relation to tumour cell metabolism remains largely unexplored. Here we show that formate, a metabolite produced by the CRC-associated bacterium Fusobacterium nucleatum, promotes CRC development. We describe molecular signatures linking CRC phenotypes with Fusobacterium abundance. Cocultures of F. nucleatum with patient-derived CRC cells display protumorigenic effects, along with a metabolic shift towards increased formate secretion and cancer glutamine metabolism. We further show that microbiome-derived formate drives CRC tumour invasion by triggering AhR signalling, while increasing cancer stemness. Finally, F. nucleatum or formate treatment in mice leads to increased tumour incidence or size, and Th17 cell expansion, which can favour proinflammatory profiles. Moving beyond observational studies, we identify formate as a gut-derived oncometabolite that is relevant for CRC progression.


Subject(s)
Colorectal Neoplasms , Gastrointestinal Microbiome , Animals , Bacteria , Colorectal Neoplasms/metabolism , Formates , Fusobacterium nucleatum , Humans , Mice , Tumor Microenvironment
9.
Proc Nutr Soc ; 80(1): 37-49, 2021 02.
Article in English | MEDLINE | ID: mdl-32238208

ABSTRACT

In recent years, the importance of the gut microbiota in human health has been revealed and many publications have highlighted its role as a key component of human physiology. Owing to the use of modern sequencing approaches, the characterisation of the microbiome in healthy individuals and in disease has demonstrated a disturbance of the microbiota, or dysbiosis, associated with pathological conditions. The microbiota establishes a symbiotic crosstalk with their host: commensal microbes benefit from the nutrient-rich environment provided by the gut and the microbiota produces hundreds of proteins and metabolites that modulate key functions of the host, including nutrient processing, maintenance of energy homoeostasis and immune system development. Many bacteria-derived metabolites originate from dietary sources. Among them, an important role has been attributed to the metabolites derived from the bacterial fermentation of dietary fibres, namely SCFA linking host nutrition to intestinal homoeostasis maintenance. SCFA are important fuels for intestinal epithelial cells (IEC) and regulate IEC functions through different mechanisms to modulate their proliferation, differentiation as well as functions of subpopulations such as enteroendocrine cells, to impact gut motility and to strengthen the gut barrier functions as well as host metabolism. Recent findings show that SCFA, and in particular butyrate, also have important intestinal and immuno-modulatory functions. In this review, we discuss the mechanisms and the impact of SCFA on gut functions and host immunity and consequently on human health.


Subject(s)
Energy Metabolism/physiology , Fatty Acids, Volatile/metabolism , Gastrointestinal Microbiome/physiology , Intestinal Mucosa/metabolism , Dysbiosis/metabolism , Homeostasis , Humans , Intestines , Nutritional Physiological Phenomena
10.
Front Immunol ; 11: 583644, 2020.
Article in English | MEDLINE | ID: mdl-33408714

ABSTRACT

Extracellular vesicles (EVs) derived from the gut microbiota are largely uncharacterized and their impacts on host intestinal physiology remain unresolved. Here, we isolated EVs from F. nucleatum for detailed characterization. Our analyses highlight the presence of the outer membrane protein porin FomA on EVs. Besides, we evaluated the impact of EVs on human intestinal epithelial cells (IECs) in a non-inflammatory context. Our results show no detrimental impact on the epithelial barrier. No internalization of EVs was observed. Moreover, we demonstrate that F. nucleatum EVs trigger innate immunity of IECs by promoting NF-κB activation via the dynamin-mediated endocytosis. The NF-κB activation was found to be TLR2-dependent yet, TLR4 was dispensable. Using competitive binding assays, we establish that FomA is involved in the NF-κB response. Taken together, our data indicate that EVs induce effects similar to those observed with whole F. nucleatum bacteria on IECs. In particular, our study highlights the role of TLR2 and FomA as major modulators of the gut epithelium immune responses to F. nucleatum.


Subject(s)
Bacterial Outer Membrane Proteins/metabolism , Extracellular Vesicles/metabolism , Fusobacterium nucleatum/metabolism , Gastrointestinal Microbiome/immunology , Toll-Like Receptor 2/metabolism , Humans , Immunity, Innate/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology
11.
Curr Opin Neurobiol ; 61: 1-9, 2020 04.
Article in English | MEDLINE | ID: mdl-31812830

ABSTRACT

The gut microbiome - the largest reservoir of microorganisms of the human body - is emerging as an important player in neurodevelopment and ageing as well as in brain diseases including stroke, Alzheimer's disease and Parkinson's disease. The growing knowledge on mediators and triggered pathways has advanced our understanding of the interactions along the gut-brain axis. Gut bacteria produce neuroactive compounds and can modulate neuronal function, plasticity and behavior. Furthermore, intestinal microorganisms impact the host's metabolism and immune status which in turn affect neuronal pathways in the enteric and central nervous systems. Here, we discuss the recent insights from human studies and animal models on the bi-directional communication along the microbiome-gut-brain axis in both acute and chronic brain diseases.


Subject(s)
Alzheimer Disease , Gastrointestinal Microbiome , Parkinson Disease , Stroke , Animals , Brain , Central Nervous System , Humans
12.
Sci Rep ; 9(1): 643, 2019 01 24.
Article in English | MEDLINE | ID: mdl-30679727

ABSTRACT

The ligand activated transcription factor, aryl hydrocarbon receptor (AhR) emerged as a critical regulator of immune and metabolic processes in the gastrointestinal tract. In the gut, a main source of AhR ligands derives from commensal bacteria. However, many of the reported microbiota-derived ligands have been restricted to indolyl metabolites. Here, by screening commensal bacteria supernatants on an AhR reporter system expressed in human intestinal epithelial cell line (IEC), we found that the short chain fatty acid (SCFA) butyrate induced AhR activity and the transcription of AhR-dependent genes in IECs. We showed that AhR ligand antagonists reduced the effects of butyrate on IEC suggesting that butyrate could act as a ligand of AhR, which was supported by the nuclear translocation of AhR induced by butyrate and in silico structural modelling. In conclusion, our findings suggest that (i) butyrate activates AhR pathway and AhR-dependent genes in human intestinal epithelial cell-lines (ii) butyrate is a potential ligand for AhR which is an original mechanism of gene regulation by SCFA.


Subject(s)
Butyrates/metabolism , Intestinal Mucosa/cytology , Receptors, Aryl Hydrocarbon/metabolism , Caco-2 Cells , HT29 Cells , Humans , Ligands , Models, Molecular , Protein Domains , Receptors, Aryl Hydrocarbon/chemistry , Receptors, G-Protein-Coupled/metabolism , Signal Transduction
13.
Sci Rep ; 8(1): 9742, 2018 06 27.
Article in English | MEDLINE | ID: mdl-29950699

ABSTRACT

The intestinal microbiota contributes to the global wellbeing of their host by their fundamental role in the induction and maintenance of a healthy immune system. Commensal bacteria shape the mucosal immune system by influencing the proportion and the activation state of anti-inflammatory regulatory T cells (Treg) by metabolites that are still only partially unravelled. Microbiota members such as Clostridiales provide a transforming growth factor ß (TGFß)-rich environment that promotes the accumulation of Treg cells in the gut. The intestinal epithelial cells (IECs) take a central part in this process, as they are a major source of TGFß1 upon bacterial colonisation. In this study, we investigated which gut commensal bacteria were able to regulate the TGFB1 human promoter in IECs using supernatants from cultured bacteria. We reported that Firmicutes and Fusobacteria supernatants were the most potent TGFB1 modulators in HT-29 cells. Furthermore, we demonstrated that butyrate was the main metabolite in bacterial supernatants accounting for TGFß1 increase. This butyrate-driven effect was independent of the G-protein coupled receptors GPR41, GPR43 and GPR109a, the transporter MCT1 as well as the transcription factors NF-κB and AP-1 present on TGFB1 promoter. Interestingly, HDAC inhibitors were inducing a similar TGFB1 increase suggesting that butyrate acted through its HDAC inhibitor properties. Finally, our results showed that SP1 was the main transcription factor mediating the HDAC inhibitor effect of butyrate on TGFB1 expression. This is, to our knowledge, the first characterisation of the mechanisms underlying TGFB1 regulation in IEC by commensal bacteria derived butyrate.


Subject(s)
Butyrates/metabolism , Epithelial Cells/metabolism , Gastrointestinal Microbiome/physiology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Intestines/cytology , Sp1 Transcription Factor/metabolism , Transforming Growth Factor beta1/metabolism , HT29 Cells , Humans , Receptors, Cell Surface/metabolism , Receptors, G-Protein-Coupled/metabolism , Transforming Growth Factor beta1/genetics
14.
Front Immunol ; 9: 2838, 2018.
Article in English | MEDLINE | ID: mdl-30619249

ABSTRACT

Commensal bacteria are crucial for the development and maintenance of a healthy immune system therefore contributing to the global well-being of their host. A wide variety of metabolites produced by commensal bacteria are influencing host health but the characterization of the multiple molecular mechanisms involved in host-microbiota interactions is still only partially unraveled. The intestinal epithelial cells (IECs) take a central part in the host-microbiota dialogue by inducing the first microbial-derived immune signals. Amongst the numerous effector molecules modulating the immune responses produced by IECs, indoleamine 2,3-dioxygenase-1 (IDO-1) is essential for gut homeostasis. IDO-1 expression is dependent on the microbiota and despites its central role, how the commensal bacteria impacts its expression is still unclear. Therefore, we investigated the impact of individual cultivable commensal bacteria on IDO-1 transcriptional expression and found that the short chain fatty acid (SCFA) butyrate was the main metabolite controlling IDO-1 expression in human primary IECs and IEC cell-lines. This butyrate-driven effect was independent of the G-protein coupled receptors GPR41, GPR43, and GPR109a and of the transcription factors SP1, AP1, and PPARγ for which binding sites were reported in the IDO-1 promoter. We demonstrated for the first time that butyrate represses IDO-1 expression by two distinct mechanisms. Firstly, butyrate decreases STAT1 expression leading to the inhibition of the IFNγ-dependent and phosphoSTAT1-driven transcription of IDO-1. In addition, we described a second mechanism by which butyrate impairs IDO-1 transcription in a STAT1-independent manner that could be attributed to its histone deacetylase (HDAC) inhibitor property. In conclusion, our results showed that IDO-1 expression is down-regulated by butyrate via a dual mechanism: the reduction of STAT1 level and the HDAC inhibitor property of SCFAs.


Subject(s)
Bacteria , Butyric Acid , Down-Regulation/immunology , Epithelial Cells , Gastrointestinal Microbiome/immunology , Gene Expression Regulation, Developmental/immunology , Indoleamine-Pyrrole 2,3,-Dioxygenase , Intestinal Mucosa , Bacteria/immunology , Bacteria/metabolism , Butyric Acid/immunology , Butyric Acid/metabolism , Caco-2 Cells , Epithelial Cells/enzymology , Epithelial Cells/immunology , Epithelial Cells/microbiology , Female , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/biosynthesis , Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Intestinal Mucosa/enzymology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Male , Middle Aged , Receptors, G-Protein-Coupled/immunology , Receptors, G-Protein-Coupled/metabolism , Transcription Factors/immunology , Transcription Factors/metabolism
16.
ISME J ; 10(2): 460-77, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26218241

ABSTRACT

Studying host-microbiota interactions are fundamental to understanding the mechanisms involved in intestinal homeostasis and inflammation. In this work, we analyzed these interactions in mice that were mono-associated with six microorganisms that are representative of inflammatory bowel disease (IBD)-associated dysbiosis: the bacteria Bacteroides thetaiotaomicron, adhesive-invasive Escherichia coli (AIEC), Ruminococcus gnavus and Roseburia intestinalis; a yeast used as a probiotic drug, Saccharomyces boulardii CNCM I-745; and another yeast, Candida albicans. Extensive ex vivo analyses including colon transcriptomics, histology, immune response, bile acid metabolism and short-chain fatty acid production were studied. We showed that B. thetaiotaomicron had the highest impact on the immune system because it was almost able to recapitulate the effects of the entire conventional microbiota and notably induced Treg pathways. Furthermore, these analyses uncovered the effects of E. coli AIEC LF82 on indoleamine 2,3-dioxygenase expression and of S. boulardii CNCM I-745 on angiogenesis. These results were confirmed in vitro in human cell lines. Finally, our results suggested that R. gnavus has major effects on metabolism, and notably on tryptophan metabolism. This work therefore reveals that microorganisms with a potential role in intestinal homeostasis and inflammation have specific impacts on the host, and it suggests several tracks to follow to understand intestinal homeostasis and IBD pathogenesis better, providing new insights to identify novel therapeutic targets.


Subject(s)
Bacteria/growth & development , Dysbiosis/microbiology , Gastrointestinal Microbiome , Inflammatory Bowel Diseases/microbiology , Intestines/microbiology , Animals , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Colon/microbiology , Disease Models, Animal , Germ-Free Life , Humans , Intestinal Mucosa/metabolism , Mice , Yeasts/genetics , Yeasts/growth & development , Yeasts/isolation & purification
SELECTION OF CITATIONS
SEARCH DETAIL