109 results on '"Blottière HM"'
Search Results
2. Short chain fatty acid propionate and the role of gb-oxidation of butyrate in rat experimental colitis
- Author
-
Xing, JH, primary, Cherbut, C., additional, Joubert, M., additional, Blottière, HM., additional, Vallette, G., additional, Laboisse, C., additional, and Galmiche, JP., additional
- Published
- 1998
- Full Text
- View/download PDF
3. Effects of sulfated polysaccharides from green seaweeds (ulvans) on the survival, proliferation and differentiation of tumoral colonic epithelial cells (HT-29 and Caco-2)
- Author
-
Bénard, C., primary, Kaeffer, B., additional, Lahaye, M., additional, Blottière, HM, additional, Galmiche, JP, additional, and Cherbut, C., additional
- Published
- 1997
- Full Text
- View/download PDF
4. Effect of butyrate on the expression of cyclin-dependent kinases and cyclin D by HT-29 epithelial cells
- Author
-
Siavoshian, S., primary, Blottière, HM, additional, Cherbut, C., additional, Kaeffer, B., additional, and Galmiche, JP, additional
- Published
- 1997
- Full Text
- View/download PDF
5. Molecular analysis of the effect of short-chain fatty acids on intestinal cell proliferation.
- Author
-
Blottière HM, Buecher B, Galmiche J, and Cherbut C
- Published
- 2003
- Full Text
- View/download PDF
6. Unraveling the benefits of Bacillus subtilis DSM 29784 poultry probiotic through its secreted metabolites: an in vitro approach.
- Author
-
Vieco-Saiz N, Prévéraud DP, Pinloche E, Morat A, Govindin P, Blottière HM, Matthieu E, Devillard E, and Consuegra J
- Subjects
- Animals, Humans, Caco-2 Cells, HT29 Cells, Fermentation, NF-kappa B metabolism, Intestinal Mucosa metabolism, Intestinal Mucosa microbiology, Intestines microbiology, Enterocytes metabolism, Niacin metabolism, Cytokines metabolism, Probiotics metabolism, Bacillus subtilis metabolism, Chickens microbiology, Gastrointestinal Microbiome physiology
- Abstract
The probiotic Bacillus subtilis 29784 (Bs29784) sustains chicken's intestinal health, enhancing animal resilience and performance through the production of the bioactive metabolites hypoxanthine (HPX), niacin (NIA), and pantothenate (PTH). Here, using enterocyte in vitro models, we determine the functional link between these metabolites and the three pillars of intestinal resilience: immune response, intestinal barrier, and microbiota. We evaluated in vitro the capacity of Bs29784 vegetative cells, spores, and metabolites to modulate global immune regulators (using HT-29-NF-κB and HT-29-AP-1 reporter cells), intestinal integrity (HT-29-MUC2 reporter cells and Caco-2 cells), and cytokine production (Caco-2 cells). Finally, we simulated intestinal fermentations using chicken's intestinal contents as inocula to determine the effect of Bs29784 metabolites on the microbiota and their fermentation profile. Bs29784 vegetative cells reduced the inflammatory response more effectively than spores, indicating that their benefit is linked to metabolic activity. To assess this hypothesis, we studied Bs29784 metabolites individually. The results showed that each metabolite had different beneficial effects. PTH and NIA reduced the activation of the pro-inflammatory pathways AP-1 and NF-κB. HPX upregulated mucin production by enhancing MUC2 expression. HPX, NIA, and PTH increased cell proliferation. PTH and HPX increased epithelial resilience to an inflammatory challenge by limiting permeability increase. In cecal fermentations, NIA increased acetate, HPX increased butyrate, whereas PTH increased acetate, butyrate, and propionate. In ileal fermentations, PTH increased butyrate. All molecules modulated microbiota, explaining the different fermentation patterns. Altogether, we show that Bs29784 influences intestinal health by acting on the three lines of resilience via its secreted metabolites., Importance: Probiotics provide beneficial metabolites to its host. Here, we describe the mode of action of a commonly used probiotic in poultry, Bs29784. By using in vitro cellular techniques and simulated chickens' intestinal model, we show the functional link between Bs29784 metabolites and the three lines of animal resilience. Indeed, both Bs29784 vegetative cells and its metabolites stimulate cellular anti-inflammatory responses, strengthen intestinal barrier, and positively modulate microbiota composition and fermentative profile. Taken together, these results strengthen our understanding of the effect of Bs29784 on its host and explain, at least partly, its positive effects on animal health, resilience, and performance., Competing Interests: The probiotic Bacillus subtilis DSM29784 is commercialized by Adisseo France SAS. N.V.-S., D.P.P., E.P., E.D., and J.C. are affiliated to Adisseo's Department of Research and Innovation. The authors declare that their affiliation with the company does not influence the results, conclusions, and analyses reported in this paper.
- Published
- 2024
- Full Text
- View/download PDF
7. Glutamate from the microbiome controls host metabolism.
- Author
-
Le Dréan G and Blottière HM
- Subjects
- Humans, Animals, Gastrointestinal Microbiome, Microbiota, Glutamic Acid metabolism
- Published
- 2024
- Full Text
- View/download PDF
8. Fusobacterium nucleatum promotes inflammatory and anti-apoptotic responses in colorectal cancer cells via ADP-heptose release and ALPK1/TIFA axis activation.
- Author
-
Martin-Gallausiaux C, Salesse L, Garcia-Weber D, Marinelli L, Beguet-Crespel F, Brochard V, Le Gléau C, Jamet A, Doré J, Blottière HM, Arrieumerlou C, and Lapaque N
- Subjects
- Humans, Fusobacterium nucleatum metabolism, Base Composition, Phylogeny, RNA, Ribosomal, 16S, Sequence Analysis, DNA, Heptoses metabolism, Tumor Microenvironment, Gastrointestinal Microbiome, Colorectal Neoplasms pathology
- 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.
- Published
- 2024
- Full Text
- View/download PDF
9. Identification of a muropeptide precursor transporter from gut microbiota and its role in preventing intestinal inflammation.
- Author
-
Liuu S, Nepelska M, Pfister H, Gamelas Magalhaes J, Chevalier G, Strozzi F, Billerey C, Maresca M, Nicoletti C, Di Pasquale E, Pechard C, Bardouillet L, Girardin SE, Boneca IG, Doré J, Blottière HM, Bonny C, Chene L, and Cultrone A
- Subjects
- Humans, Mice, Animals, Peptidoglycan metabolism, Intestines pathology, Inflammation metabolism, Membrane Transport Proteins metabolism, Anti-Inflammatory Agents metabolism, Dextran Sulfate, Disease Models, Animal, Colon metabolism, Mice, Inbred C57BL, Gastrointestinal Microbiome, Colitis metabolism
- Abstract
The gut microbiota is a considerable source of biologically active compounds that can promote intestinal homeostasis and improve immune responses. Here, we used large expression libraries of cloned metagenomic DNA to identify compounds able to sustain an anti-inflammatory reaction on host cells. Starting with a screen for NF-κB activation, we have identified overlapping clones harbouring a heterodimeric ATP-binding cassette (ABC)-transporter from a Firmicutes. Extensive purification of the clone's supernatant demonstrates that the ABC-transporter allows for the efficient extracellular accumulation of three muropeptide precursor, with anti-inflammatory properties. They induce IL-10 secretion from human monocyte-derived dendritic cells and proved effective in reducing AIEC LF82 epithelial damage and IL-8 secretion in human intestinal resections. In addition, treatment with supernatants containing the muropeptide precursor reduces body weight loss and improves histological parameters in Dextran Sulfate Sodium (DSS)-treated mice. Until now, the source of peptidoglycan fragments was shown to come from the natural turnover of the peptidoglycan layer by endogenous peptidoglycan hydrolases. This is a report showing an ABC-transporter as a natural source of secreted muropeptide precursor and as an indirect player in epithelial barrier strengthening. The mechanism described here might represent an important component of the host immune homeostasis., Competing Interests: Competing interests statement:M.N., J.D., H.M.B., A.C., and C. Bonny are the inventors of a patent application protecting the heterodimeric transporter (WO 2021/148661). The other authors have no conflicts of interest to declare aside from the fact that several of the authors are employees of private companies.
- Published
- 2023
- Full Text
- View/download PDF
10. An Insight into Functional Metagenomics: A High-Throughput Approach to Decipher Food-Microbiota-Host Interactions in the Human Gut.
- Author
-
Mathieu E, Léjard V, Ezzine C, Govindin P, Morat A, Giat M, Lapaque N, Doré J, and Blottière HM
- Subjects
- Humans, Host Microbial Interactions, Metagenomics methods, Metagenome, Microbiota, Gastrointestinal Microbiome
- Abstract
Our understanding of the symbiotic relationship between the microbiota and its host has constantly evolved since our understanding that the "self" was not only defined by our genetic patrimony but also by the genomes of bugs living in us. The first culture-based methods highlighted the important functions of the microbiota. However, these methods had strong limitations and did not allow for a full understanding of the complex relationships that occur at the interface between the microbiota and the host. The recent development of metagenomic approaches has been a groundbreaking step towards this understanding. Its use has provided new insights and perspectives. In the present chapter, we will describe the advances of functional metagenomics to decipher food-microbiota and host-microbiota interactions. This powerful high-throughput approach allows for the assessment of the microbiota as a whole (including non-cultured bacteria) and enabled the discovery of new signaling pathways and functions involved in the crosstalk between food, the gut microbiota and its host. We will present the pipeline and highlight the most important studies that helped to develop the field. To conclude, we will emphasize the most recent developments and hot topics in functional metagenomics., Competing Interests: The authors declare no conflicts of interest.
- Published
- 2023
- Full Text
- View/download PDF
11. The bacterial species profiles of the lingual and salivary microbiota differ with basic tastes sensitivity in human.
- Author
-
Licandro H, Truntzer C, Fromentin S, Morabito C, Quinquis B, Pons N, Martin C, Blottière HM, and Neyraud E
- Subjects
- Humans, Taste, Saliva, Tongue, Taste Perception, Microbiota
- Abstract
Taste perception is crucial and impairments, which can be linked to pathologies, can lead to eating disorders. It is triggered by taste compounds stimulating receptors located on the tongue. However, the tongue is covered by a film containing saliva and microorganisms suspected to modulate the taste receptor environment. The present study aimed to elucidate the links between taste sensitivity (sweetness, sourness, bitterness, saltiness, umami) and the salivary as well as the tongue microbiota using shotgun metagenomics. 109 bacterial species were correlated with at least one taste. Interestingly, when a species was correlated with at least two tastes, the correlations were unidirectional, indicating a putative global implication. Some Streptococcus, SR1 and Rickenellaceae species correlated with five tastes. When comparing both ecosystems, saliva appears to be a better taste predictor than tongue. This work shows the implication of the oral microbiota in taste and exhibits specificities depending on the ecosystem considered., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
12. α-Synuclein expression in response to bacterial ligands and metabolites in gut enteroendocrine cells: an in vitro proof of concept study.
- Author
-
Hurley MJ, Menozzi E, Koletsi S, Bates R, Gegg ME, Chau KY, Blottière HM, Macnaughtan J, and Schapira AHV
- Abstract
Caudo-rostral migration of pathological forms of α-synuclein from the gut to the brain is proposed as an early feature in Parkinson's disease pathogenesis, but the underlying mechanisms remain unknown. Intestinal epithelial enteroendocrine cells sense and respond to numerous luminal signals, including bacterial factors, and transmit this information to the brain via the enteric nervous system and vagus nerve. There is evidence that gut bacteria composition and their metabolites change in Parkinson's disease patients, and these alterations can trigger α-synuclein pathology in animal models of the disorder. Here, we investigated the effect of toll-like receptor and free fatty acid receptor agonists on the intracellular level of α-synuclein and its release using mouse secretin tumour cell line 1 enteroendocrine cells. Secretin tumour cell line 1 enteroendocrine cells were treated for 24 or 48 h with toll-like receptor agonists (toll-like receptor 4 selective lipopolysaccharide; toll-like receptor 2 selective Pam3CysSerLys4) and the free fatty acid receptor 2/3 agonists butyrate, propionate and acetate. The effect of selective receptor antagonists on the agonists' effects after 24 hours was also investigated. The level of α-synuclein protein was measured in cell lysates and cell culture media by western blot and enzyme-linked immunosorbent assay. The level of α-synuclein and tumour necrosis factor messenger RNA was measured by quantitative reverse transcription real-time polymerase chain reaction. Stimulation of secretin tumour cell line 1 enteroendocrine cells for 24 and 48 hours with toll-like receptor and free fatty acid receptor agonists significantly increased the amount of intracellular α-synuclein and the release of α-synuclein from the cells into the culture medium. Both effects were significantly reduced by antagonists selective for each receptor. Toll-like receptor and free fatty acid receptor agonists also significantly increased tumour necrosis factor transcription, and this was effectively inhibited by corresponding antagonists. Elevated intracellular α-synuclein increases the likelihood of aggregation and conversion to toxic forms. Factors derived from bacteria induce α-synuclein accumulation in secretin tumour cell line 1 enteroendocrine cells. Here, we provide support for a mechanism by which exposure of enteroendocrine cells to specific bacterial factors found in Parkinson's disease gut dysbiosis might facilitate accumulation of α-synuclein pathology in the gut., Competing Interests: The authors report no competing interests or relevant disclosures., (© The Author(s) 2023. Published by Oxford University Press on behalf of the Guarantors of Brain.)
- Published
- 2023
- Full Text
- View/download PDF
13. Adlercreutzia equolifaciens Is an Anti-Inflammatory Commensal Bacterium with Decreased Abundance in Gut Microbiota of Patients with Metabolic Liver Disease.
- Author
-
Oñate FP, Chamignon C, Burz SD, Lapaque N, Monnoye M, Philippe C, Bredel M, Chêne L, Farin W, Paillarse JM, Boursier J, Ratziu V, Mousset PY, Doré J, Gérard P, and Blottière HM
- Subjects
- Animals, Mice, Dysbiosis microbiology, Liver metabolism, Anti-Inflammatory Agents pharmacology, Anti-Inflammatory Agents therapeutic use, Anti-Inflammatory Agents metabolism, Non-alcoholic Fatty Liver Disease metabolism, Gastrointestinal Microbiome, Metabolic Diseases metabolism, Liver Neoplasms metabolism
- Abstract
Non-alcoholic fatty liver disease (NAFLD) affects about 20-40% of the adult population in high-income countries and is now a leading indication for liver transplantation and can lead to hepatocellular carcinoma. The link between gut microbiota dysbiosis and NAFLD is now clearly established. Through analyses of the gut microbiota with shotgun metagenomics, we observe that compared to healthy controls, Adlercreutzia equolifaciens is depleted in patients with liver diseases such as NAFLD. Its abundance also decreases as the disease progresses and eventually disappears in the last stages indicating a strong association with disease severity. Moreover, we show that A. equolifaciens possesses anti-inflammatory properties, both in vitro and in vivo in a humanized mouse model of NAFLD. Therefore, our results demonstrate a link between NAFLD and the severity of liver disease and the presence of A. equolifaciens and its anti-inflammatory actions. Counterbalancing dysbiosis with this bacterium may be a promising live biotherapeutic strategy for liver diseases.
- Published
- 2023
- Full Text
- View/download PDF
14. Cross talk between Paneth and tuft cells drives dysbiosis and inflammation in the gut mucosa.
- Author
-
Coutry N, Nguyen J, Soualhi S, Gerbe F, Meslier V, Dardalhon V, Almeida M, Quinquis B, Thirion F, Herbert F, Gasmi I, Lamrani A, Giordano A, Cesses P, Garnier L, Thirard S, Greuet D, Cazevieille C, Bernex F, Bressuire C, Winton D, Matsumoto I, Blottière HM, Taylor N, and Jay P
- Subjects
- Humans, Inflammation, Paneth Cells, Succinates, Succinic Acid, Dysbiosis, Mucous Membrane
- Abstract
Gut microbiota imbalance (dysbiosis) is increasingly associated with pathological conditions, both within and outside the gastrointestinal tract. Intestinal Paneth cells are considered to be guardians of the gut microbiota, but the events linking Paneth cell dysfunction with dysbiosis remain unclear. We report a three-step mechanism for dysbiosis initiation. Initial alterations in Paneth cells, as frequently observed in obese and inflammatorybowel diseases patients, cause a mild remodeling of microbiota, with amplification of succinate-producing species. SucnR1-dependent activation of epithelial tuft cells triggers a type 2 immune response that, in turn, aggravates the Paneth cell defaults, promoting dysbiosis and chronic inflammation. We thus reveal a function of tuft cells in promoting dysbiosis following Paneth cell deficiency and an unappreciated essential role of Paneth cells in maintaining a balanced microbiota to prevent inappropriate activation of tuft cells and deleterious dysbiosis. This succinate-tuft cell inflammation circuit may also contribute to the chronic dysbiosis observed in patients.
- Published
- 2023
- Full Text
- View/download PDF
15. Roseburia, a decreased bacterial taxon in the gut microbiota of patients suffering from anorexia nervosa.
- Author
-
Mondot S, Lachkar L, Doré J, Blottière HM, and Hanachi M
- Subjects
- Bacteria genetics, Cathartics, Feces microbiology, Humans, RNA, Ribosomal, 16S genetics, Anorexia Nervosa complications, Gastrointestinal Microbiome
- Abstract
Anorexia nervosa (AN) is a severe eating disorder which can lead to malnutrition and life threatening complications with high mortality rates. We designed our analysis to identify gut microbial taxa differentially abundant between AN and HC across different 16S rRNA gene datasets. We identified a reduced abundance, diversity and richness of Roseburia genus in the microbiota of patients with AN. Cares leading to partial recovery of patients with AN during hospitalization did not restore Roseburia to the levels of HC. AN dietary habit, either purgative or restrictive, did not affect Roseburia abundance. Roseburia genus and related species abundance were correlated with different health host metabolic markers. Roseburia species are key functional taxa in the human gut microbiome. Low gut Roseburia levels have been linked with other human pathologies. Our study highlights Roseburia species as a major decreased component in the gut of patients with AN., (© 2022. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2022
- Full Text
- View/download PDF
16. Neonatal Programming of Microbiota Composition: A Plausible Idea That Is Not Supported by the Evidence.
- Author
-
Michel C and Blottière HM
- Abstract
Underpinning the theory "developmental origins of health and disease" (DOHaD), evidence is accumulating to suggest that the risks of adult disease are in part programmed by exposure to environmental factors during the highly plastic "first 1,000 days of life" period. An elucidation of the mechanisms involved in this programming is challenging as it would help developing new strategies to promote adult health. The intestinal microbiome is proposed as a long-lasting memory of the neonatal environment. This proposal is supported by indisputable findings such as the concomitance of microbiota assembly and the first 1,000-day period, the influence of perinatal conditions on microbiota composition, and the impact of microbiota composition on host physiology, and is based on the widely held but unconfirmed view that the microbiota is long-lastingly shaped early in life. In this review, we examine the plausibility of the gut microbiota being programmed by the neonatal environment and evaluate the evidence for its validity. We highlight that the capacity of the pioneer bacteria to control the implantation of subsequent bacteria is supported by both theoretical principles and statistical associations, but remains to be demonstrated experimentally. In addition, our critical review of the literature on the long-term repercussions of selected neonatal modulations of the gut microbiota indicates that sustained programming of the microbiota composition by neonatal events is unlikely. This does not exclude the microbiota having a role in DOHaD due to a possible interaction with tissue and organ development during the critical windows of neonatal life., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Michel and Blottière.)
- Published
- 2022
- Full Text
- View/download PDF
17. Akkermansia muciniphila upregulates genes involved in maintaining the intestinal barrier function via ADP-heptose-dependent activation of the ALPK1/TIFA pathway.
- Author
-
Martin-Gallausiaux C, Garcia-Weber D, Lashermes A, Larraufie P, Marinelli L, Teixeira V, Rolland A, Béguet-Crespel F, Brochard V, Quatremare T, Jamet A, Doré J, Gray-Owen SD, Blottière HM, Arrieumerlou C, and Lapaque N
- Subjects
- Adenosine Diphosphate, Akkermansia, Heptoses, Immunity, Innate, TNF Receptor-Associated Factor 6, Verrucomicrobia, Gastrointestinal Microbiome, NF-kappa B
- 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.
- Published
- 2022
- Full Text
- View/download PDF
18. SCFA: mechanisms and functional importance in the gut.
- Author
-
Martin-Gallausiaux C, Marinelli L, Blottière HM, Larraufie P, and Lapaque N
- Subjects
- Dysbiosis metabolism, Homeostasis, Humans, Intestines, Nutritional Physiological Phenomena, Energy Metabolism physiology, Fatty Acids, Volatile metabolism, Gastrointestinal Microbiome physiology, Intestinal Mucosa metabolism
- 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.
- Published
- 2021
- Full Text
- View/download PDF
19. Fecal Microbiota Transplant from Human to Mice Gives Insights into the Role of the Gut Microbiota in Non-Alcoholic Fatty Liver Disease (NAFLD).
- Author
-
Burz SD, Monnoye M, Philippe C, Farin W, Ratziu V, Strozzi F, Paillarse JM, Chêne L, Blottière HM, and Gérard P
- Abstract
Non-alcoholic fatty liver diseases (NAFLD) are associated with changes in the composition and metabolic activities of the gut microbiota. However, the causal role played by the gut microbiota in individual susceptibility to NAFLD and particularly at its early stage is still unclear. In this context, we transplanted the microbiota from a patient with fatty liver (NAFL) and from a healthy individual to two groups of mice. We first showed that the microbiota composition in recipient mice resembled the microbiota composition of their respective human donor. Following administration of a high-fructose, high-fat diet, mice that received the human NAFL microbiota (NAFLR) gained more weight and had a higher liver triglycerides level and higher plasma LDL cholesterol than mice that received the human healthy microbiota (HR). Metabolomic analyses revealed that it was associated with lower and higher plasma levels of glycine and 3-Indolepropionic acid in NAFLR mice, respectively. Moreover, several bacterial genera and OTUs were identified as differently represented in the NAFLR and HR microbiota and therefore potentially responsible for the different phenotypes observed. Altogether, our results confirm that the gut bacteria play a role in obesity and steatosis development and that targeting the gut microbiota may be a preventive or therapeutic strategy in NAFLD management.
- Published
- 2021
- Full Text
- View/download PDF
20. Alternative stable states in the intestinal ecosystem: proof of concept in a rat model and a perspective of therapeutic implications.
- Author
-
Van de Guchte M, Burz SD, Cadiou J, Wu J, Mondot S, Blottière HM, and Doré J
- Subjects
- Animals, Dextran Sulfate pharmacology, Inflammation chemically induced, Inflammation microbiology, Intestines drug effects, Intestines pathology, Male, RNA, Ribosomal, 16S genetics, Rats, Gastrointestinal Microbiome drug effects, Gastrointestinal Microbiome genetics, Intestines microbiology
- Abstract
Background: Chronic immune-mediated diseases are rapidly expanding and notoriously difficult to cure. Altered relatively stable intestinal microbiota configurations are associated with several of these diseases, and with a possible pre-disease condition (more susceptible to disease development) of the host-microbiota ecosystem. These observations are reminiscent of the behavior of an ecosystem with alternative stable states (different stable configurations that can exist under identical external conditions), and we recently postulated that health, pre-disease and disease represent such alternative states. Here, our aim was to examine if alternative stable states indeed exist in the intestinal ecosystem., Results: Rats were exposed to varying concentrations of DSS in order to create a wide range of mildly inflammatory conditions, in a context of diet-induced low microbiota diversity. The consequences for the intestinal microbiota were traced by 16S rRNA gene profiling over time, and inflammation of the distal colon was evaluated at sacrifice, 45 days after the last DSS treatment. The results provide the first formal experimental proof for the existence of alternative stable states in the rat intestinal ecosystem, taking both microbiota and host inflammatory status into consideration. The alternative states are host-microbiota ecosystem states rather than independent and dissociated microbiota and host states, and inflammation can prompt stable state-transition. Based on these results, we propose a conceptual model providing new insights in the interplay between host inflammatory status and microbiota status. These new insights call for innovative therapeutic strategies to cure (pre-)disease., Conclusions: We provide proof of concept showing the existence of alternative stable states in the rat intestinal ecosystem. We further propose a model which, if validated in humans, will support innovative diagnosis, therapeutic strategy, and monitoring in the treatment of chronic inflammatory conditions. This model provides a strong rationale for the application of combinatorial therapeutic strategies, targeting host and microbiota rather than only one of the two in chronic immune-mediated diseases. Video Abstract.
- Published
- 2020
- Full Text
- View/download PDF
21. Impact of pemetrexed chemotherapy on the gut microbiota and intestinal inflammation of patient-lung-derived tumor xenograft (PDX) mouse models.
- Author
-
Pensec C, Gillaizeau F, Guenot D, Bessard A, Carton T, Leuillet S, Campone M, Neunlist M, Blottière HM, and Le Vacon F
- Subjects
- Animals, Carcinoma, Non-Small-Cell Lung pathology, Disease Models, Animal, Female, Heterografts, Inflammation, Intestinal Mucosa drug effects, Intestinal Mucosa pathology, Lung Neoplasms pathology, Mice, SCID, Specific Pathogen-Free Organisms, Antineoplastic Agents adverse effects, Carcinoma, Non-Small-Cell Lung drug therapy, Carcinoma, Non-Small-Cell Lung microbiology, Gastrointestinal Microbiome, Intestinal Mucosa microbiology, Lung Neoplasms drug therapy, Lung Neoplasms microbiology, Neoplasm Transplantation, Pemetrexed adverse effects
- Abstract
Chemotherapy remains the gold standard for advanced cancer. Pemetrexed, a chemotherapeutic agent used in non-small cell lung cancer, can induce significant side effects in patients. Although microbiota's role in the efficacy and/or toxicity of chemotherapy agents has been demonstrated, the impacts of pemetrexed on the gut microbiota and on gastrointestinal inflammation remain unknown. The objective of this study was to evaluate the impact of pemetrexed and the tumor graft on the gut microbiota composition in immunodeficient mice. The faecal microbiota composition was studied with metabarcoding before, 24-h and one week after treatment. The colon epithelial barrier integrity was evaluated by histological examination, intestinal permeability measurement, and selected cytokines quantification. The tumor graft induced some variations in the microbiota composition. Pemetrexed further increased the relative abundance of Enterobacteriaceae and 3 families from the Firmicutes phylum: Enterococcaceae, Lactobacillaceae and Streptococcaceae. Pemetrexed also significantly altered the epithelial barrier integrity, which was associated with early inflammation. This pilot study shows that the association of a lung tumor graft with pemetrexed causes an alteration in the microbiota composition. Such information increases our knowledge about the impact of chemotherapy on the microbiota, which could help to minimize side effects and improve therapeutic effectiveness in the future.
- Published
- 2020
- Full Text
- View/download PDF
22. Microbiota-gut brain axis involvement in neuropsychiatric disorders.
- Author
-
Iannone LF, Preda A, Blottière HM, Clarke G, Albani D, Belcastro V, Carotenuto M, Cattaneo A, Citraro R, Ferraris C, Ronchi F, Luongo G, Santocchi E, Guiducci L, Baldelli P, Iannetti P, Pedersen S, Petretto A, Provasi S, Selmer K, Spalice A, Tagliabue A, Verrotti A, Segata N, Zimmermann J, Minetti C, Mainardi P, Giordano C, Sisodiya S, Zara F, Russo E, and Striano P
- Subjects
- Humans, Central Nervous System Diseases microbiology, Gastrointestinal Microbiome, Mental Disorders microbiology
- Abstract
Introduction : The microbiota-gut brain (MGB) axis is the bidirectional communication between the intestinal microbiota and the brain. An increasing body of preclinical and clinical evidence has revealed that the gut microbial ecosystem can affect neuropsychiatric health. However, there is still a need of further studies to elucidate the complex gene-environment interactions and the role of the MGB axis in neuropsychiatric diseases, with the aim of identifying biomarkers and new therapeutic targets, to allow early diagnosis and improving treatments. Areas covered : To review the role of MGB axis in neuropsychiatric disorders, prediction and prevention of disease through exploitation, integration, and combination of data from existing gut microbiome/microbiota projects and appropriate other International '-Omics' studies. The authors also evaluated the new technological advances to investigate and modulate, through nutritional and other interventions, the gut microbiota. Expert opinion : The clinical studies have documented an association between alterations in gut microbiota composition and/or function, whereas the preclinical studies support a role for the gut microbiota in impacting behaviors which are of relevance to psychiatry and other central nervous system (CNS) disorders. Targeting MGB axis could be an additional approach for treating CNS disorders and all conditions in which alterations of the gut microbiota are involved.
- Published
- 2019
- Full Text
- View/download PDF
23. A Guide for Ex Vivo Handling and Storage of Stool Samples Intended for Fecal Microbiota Transplantation.
- Author
-
Burz SD, Abraham AL, Fonseca F, David O, Chapron A, Béguet-Crespel F, Cénard S, Le Roux K, Patrascu O, Levenez F, Schwintner C, Blottière HM, Béra-Maillet C, Lepage P, Doré J, and Juste C
- Subjects
- Cryoprotective Agents, Humans, Polysaccharides, Trehalose, Fecal Microbiota Transplantation, Feces, Guidelines as Topic, Specimen Handling methods
- Abstract
Owing to the growing recognition of the gut microbiota as a main partner of human health, we are expecting that the number of indications for fecal microbiota transplantation (FMT) will increase. Thus, there is an urgent need for standardization of the entire process of fecal transplant production. This study provides a complete standardized procedure to prepare and store live and ready-to-use transplants that meet the standard requirements of good practices to applied use in pharmaceutical industry. We show that, if time before transformation to transplants would exceed 24 hours, fresh samples should not be exposed to temperatures above 20 °C, and refrigeration at 4 °C can be a safe solution. Oxygen-free atmosphere was not necessary and simply removing air above collected samples was sufficient to preserve viability. Transplants prepared in maltodextrin-trehalose solutions, stored in a -80 °C standard freezer and then rapidly thawed at 37 °C, retained the best revivification potential as proven by 16S rRNA profiles, metabolomic fingerprints, and flow cytometry assays over a 3-month observation period. Maltodextrin-trehalose containing cryoprotectants were also efficient in preserving viability of lyophilized transplants, either in their crude or purified form, an option that can be attractive for fecal transplant biobanking and oral formulation.
- Published
- 2019
- Full Text
- View/download PDF
24. Fructose malabsorption induces cholecystokinin expression in the ileum and cecum by changing microbiota composition and metabolism.
- Author
-
Zhang X, Grosfeld A, Williams E, Vasiliauskas D, Barretto S, Smith L, Mariadassou M, Philippe C, Devime F, Melchior C, Gourcerol G, Dourmap N, Lapaque N, Larraufie P, Blottière HM, Herberden C, Gerard P, Rehfeld JF, Ferraris RP, Fritton JC, Ellero-Simatos S, and Douard V
- Subjects
- Animals, Cecum drug effects, Cell Line, Fructokinases genetics, Fructokinases metabolism, Fructose administration & dosage, Gene Expression Regulation drug effects, Humans, Ileum drug effects, Mice, Mice, Knockout, Cecum metabolism, Cholecystokinin metabolism, Fructose metabolism, Fructose pharmacology, Gastrointestinal Microbiome drug effects, Ileum metabolism
- Abstract
Current fructose consumption levels often overwhelm the intestinal capacity to absorb fructose. We investigated the impact of fructose malabsorption on intestinal endocrine function and addressed the role of the microbiota in this process. To answer this question, a mouse model of moderate fructose malabsorption [ketohexokinase mutant (KHK)
-/- ] and wild-type (WT) littermate mice were used and received a 20%-fructose (KHK-F and WT-F) or 20%-glucose diet. Cholecystokinin ( Cck ) mRNA and protein expression in the ileum and cecum, as well as preproglucagon ( Gcg ) and neurotensin ( Nts ) mRNA expression in the cecum, increased in KHK-F mice. In KHK-F mice, triple-label immunohistochemistry showed major up-regulation of CCK in enteroendocrine cells (EECs) that were glucagon-like peptide-1 (GLP-1)+ /Peptide YY (PYY- ) in the ileum and colon and GLP-1- /PYY- in the cecum. The cecal microbiota composition was drastically modified in the KHK-F in association with an increase in glucose, propionate, succinate, and lactate concentrations. Antibiotic treatment abolished fructose malabsorption-dependent induction of cecal Cck mRNA expression and, in mouse GLUTag and human NCI-H716 cells, Cck mRNA expression levels increased in response to propionate, both suggesting a microbiota-dependent process. Fructose reaching the lower intestine can modify the composition and metabolism of the microbiota, thereby stimulating the production of CCK from the EECs possibly in response to propionate.-Zhang, X., Grosfeld, A., Williams, E., Vasiliauskas, D., Barretto, S., Smith, L., Mariadassou, M., Philippe, C., Devime, F., Melchior, C., Gourcerol, G., Dourmap, N., Lapaque, N., Larraufie, P., Blottière, H. M., Herberden, C., Gerard, P., Rehfeld, J. F., Ferraris, R. P., Fritton, J. C., Ellero-Simatos, S., Douard, V. Fructose malabsorption induces cholecystokinin expression in the ileum and cecum by changing microbiota composition and metabolism.- Published
- 2019
- Full Text
- View/download PDF
25. Identification of the novel role of butyrate as AhR ligand in human intestinal epithelial cells.
- Author
-
Marinelli L, Martin-Gallausiaux C, Bourhis JM, Béguet-Crespel F, Blottière HM, and Lapaque N
- Subjects
- Caco-2 Cells, HT29 Cells, Humans, Ligands, Models, Molecular, Protein Domains, Receptors, Aryl Hydrocarbon chemistry, Receptors, G-Protein-Coupled metabolism, Signal Transduction, Butyrates metabolism, Intestinal Mucosa cytology, Receptors, Aryl Hydrocarbon metabolism
- 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.
- Published
- 2019
- Full Text
- View/download PDF
26. Prediction of the intestinal resistome by a three-dimensional structure-based method.
- Author
-
Ruppé E, Ghozlane A, Tap J, Pons N, Alvarez AS, Maziers N, Cuesta T, Hernando-Amado S, Clares I, Martínez JL, Coque TM, Baquero F, Lanza VF, Máiz L, Goulenok T, de Lastours V, Amor N, Fantin B, Wieder I, Andremont A, van Schaik W, Rogers M, Zhang X, Willems RJL, de Brevern AG, Batto JM, Blottière HM, Léonard P, Léjard V, Letur A, Levenez F, Weiszer K, Haimet F, Doré J, Kennedy SP, and Ehrlich SD
- Subjects
- Bacteria classification, Bacteria genetics, Bacterial Proteins chemistry, Bacterial Proteins genetics, beta-Lactamases chemistry, beta-Lactamases genetics, Anti-Bacterial Agents pharmacology, Bacteria drug effects, Drug Resistance, Bacterial genetics, Gastrointestinal Microbiome genetics, Intestines microbiology, Protein Conformation
- Abstract
The intestinal microbiota is considered to be a major reservoir of antibiotic resistance determinants (ARDs) that could potentially be transferred to bacterial pathogens via mobile genetic elements. Yet, this assumption is poorly supported by empirical evidence due to the distant homologies between known ARDs (mostly from culturable bacteria) and ARDs from the intestinal microbiota. Consequently, an accurate census of intestinal ARDs (that is, the intestinal resistome) has not yet been fully determined. For this purpose, we developed and validated an annotation method (called pairwise comparative modelling) on the basis of a three-dimensional structure (homology comparative modelling), leading to the prediction of 6,095 ARDs in a catalogue of 3.9 million proteins from the human intestinal microbiota. We found that the majority of predicted ARDs (pdARDs) were distantly related to known ARDs (mean amino acid identity 29.8%) and found little evidence supporting their transfer between species. According to the composition of their resistome, we were able to cluster subjects from the MetaHIT cohort (n = 663) into six resistotypes that were connected to the previously described enterotypes. Finally, we found that the relative abundance of pdARDs was positively associated with gene richness, but not when subjects were exposed to antibiotics. Altogether, our results indicate that the majority of intestinal microbiota ARDs can be considered intrinsic to the dominant commensal microbiota and that these genes are rarely shared with bacterial pathogens.
- Published
- 2019
- Full Text
- View/download PDF
27. Butyrate Produced by Commensal Bacteria Down-Regulates Indolamine 2,3-Dioxygenase 1 ( IDO-1 ) Expression via a Dual Mechanism in Human Intestinal Epithelial Cells.
- Author
-
Martin-Gallausiaux C, Larraufie P, Jarry A, Béguet-Crespel F, Marinelli L, Ledue F, Reimann F, Blottière HM, and Lapaque N
- Subjects
- Caco-2 Cells, Female, Humans, Interferon-gamma biosynthesis, Interferon-gamma immunology, Male, Middle Aged, Receptors, G-Protein-Coupled immunology, Receptors, G-Protein-Coupled metabolism, Transcription Factors immunology, Transcription Factors metabolism, Bacteria immunology, Bacteria metabolism, Butyric Acid immunology, Butyric Acid metabolism, Down-Regulation immunology, Epithelial Cells enzymology, Epithelial Cells immunology, Epithelial Cells microbiology, Gastrointestinal Microbiome immunology, Gene Expression Regulation, Developmental immunology, Indoleamine-Pyrrole 2,3,-Dioxygenase biosynthesis, Indoleamine-Pyrrole 2,3,-Dioxygenase immunology, Intestinal Mucosa enzymology, Intestinal Mucosa immunology, Intestinal Mucosa microbiology
- 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.
- Published
- 2018
- Full Text
- View/download PDF
28. Butyrate produced by gut commensal bacteria activates TGF-beta1 expression through the transcription factor SP1 in human intestinal epithelial cells.
- Author
-
Martin-Gallausiaux C, Béguet-Crespel F, Marinelli L, Jamet A, Ledue F, Blottière HM, and Lapaque N
- Subjects
- HT29 Cells, Humans, Receptors, Cell Surface metabolism, Receptors, G-Protein-Coupled metabolism, Transforming Growth Factor beta1 genetics, 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
- 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 β (TGFβ)-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 TGFβ1 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 TGFβ1 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.
- Published
- 2018
- Full Text
- View/download PDF
29. Humans as holobionts: implications for prevention and therapy.
- Author
-
van de Guchte M, Blottière HM, and Doré J
- Subjects
- Bacteria classification, Dysbiosis microbiology, Host Microbial Interactions physiology, Humans, Inflammation microbiology, Inflammatory Bowel Diseases microbiology, Obesity microbiology, Bacteria immunology, Bacterial Physiological Phenomena immunology, Gastrointestinal Microbiome immunology, Host Microbial Interactions immunology, Symbiosis physiology
- Abstract
The human gut microbiota is increasingly recognized for its important or even decisive role in health. As it becomes clear that microbiota and host mutually affect and depend on each other in an intimate relationship, a holistic view of the gut microbiota-host association imposes itself. Ideally, a stable state of equilibrium, homeostasis, is maintained and serves health, but signs are that perturbation of this equilibrium beyond the limits of resilience can propel the system into an alternative stable state, a pre-disease state, more susceptible to the development of chronic diseases. The microbiota-host equilibrium of a large and growing proportion of individuals in Western society may represent such a pre-disease state and explain the explosive development of chronic diseases such as inflammatory bowel disease, obesity, and other inflammatory diseases. These diseases themselves represent other alternative stable states again and are therefore hard to cure. The holistic view of the microbiota-host association where feedback loops between microbiota and host are thought to maintain the system in a stable state-be it a healthy, pre-disease, or disease state-implies that integrated approaches, addressing host processes and microbiota, should be used to treat or prevent (pre-)disease.
- Published
- 2018
- Full Text
- View/download PDF
30. The gut bacterium and pathobiont Bacteroides vulgatus activates NF-κB in a human gut epithelial cell line in a strain and growth phase dependent manner.
- Author
-
Ó Cuív P, de Wouters T, Giri R, Mondot S, Smith WJ, Blottière HM, Begun J, and Morrison M
- Subjects
- Cell Line, Chemokine CCL2 biosynthesis, Chemokine CXCL10 biosynthesis, Gene Expression, Humans, Interleukin-6 biosynthesis, Interleukin-8 biosynthesis, Protein Transport, Up-Regulation, Bacteroides immunology, Epithelial Cells immunology, Epithelial Cells microbiology, Gastrointestinal Tract immunology, Gastrointestinal Tract microbiology, Host-Pathogen Interactions, NF-kappa B metabolism
- Abstract
The gut microbiota is increasingly implicated in the pathogenesis of Crohn's disease (CD) and ulcerative colitis (UC) although the identity of the bacteria that underpin these diseases has remained elusive. The pathobiont Bacteroides vulgatus has been associated with both diseases although relatively little is known about how its growth and functional activity might drive the host inflammatory response. We identified an ATP Binding Cassette (ABC) export system and lipoprotein in B. vulgatus ATCC 8482 and B. vulgatus PC510 that displayed significant sequence similarity to an NF-κB immunomodulatory regulon previously identified on a CD-derived metagenomic fosmid clone. Interestingly, the ABC export system was specifically enriched in CD subjects suggesting that it may be important for colonization and persistence in the CD gut environment. Both B. vulgatus ATCC 8482 and PC510 activated NF-κB in a strain and growth phase specific manner in a HT-29/kb-seap-25 enterocyte like cell line. B. vulgatus ATCC 8482 also activated NF-κB in a Caco-2-NF-κBluc enterocyte like and an LS174T-NF-κBluc goblet cell like cell lines, and induced NF-κB-p65 subunit nuclear translocation and IL-6, IL-8, CXCL-10 and MCP-1 gene expression. Despite this, NF-κB activation was not coincident with maximal expression of the ABC exporter or lipoprotein in B. vulgatus PC510 suggesting that the regulon may be necessary but not sufficient for the immunomodulatory effects., (Crown Copyright © 2017. Published by Elsevier Ltd. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
31. The Enterococcus faecalis virulence factor ElrA interacts with the human Four-and-a-Half LIM Domains Protein 2.
- Author
-
Jamet A, Dervyn R, Lapaque N, Bugli F, Perez-Cortez NG, Blottière HM, Twizere JC, Sanguinetti M, Posteraro B, Serror P, and Maguin E
- Subjects
- Animals, Bacterial Proteins chemistry, Bacterial Proteins genetics, Cell Line, Enterococcus faecalis pathogenicity, Female, Humans, LIM-Homeodomain Proteins chemistry, Mice, Muscle Proteins chemistry, Protein Binding, Protein Interaction Domains and Motifs, Transcription Factors chemistry, Virulence Factors chemistry, Virulence Factors genetics, Bacterial Proteins metabolism, Enterococcus faecalis physiology, Gram-Positive Bacterial Infections metabolism, Gram-Positive Bacterial Infections microbiology, Host-Pathogen Interactions, LIM-Homeodomain Proteins metabolism, Muscle Proteins metabolism, Transcription Factors metabolism, Virulence Factors metabolism
- Abstract
The commensal bacterium Enterococcus faecalis is a common cause of nosocomial infections worldwide. The increasing prevalence of multi-antibiotic resistant E. faecalis strains reinforces this public health concern. Despite numerous studies highlighting several pathology-related genetic traits, the molecular mechanisms of E. faecalis virulence remain poorly understood. In this work, we studied 23 bacterial proteins that could be considered as virulence factors or involved in the Enterococcus interaction with the host. We systematically tested their interactions with human proteins using the Human ORFeome library, a set of 12,212 human ORFs, in yeast. Among the thousands of tested interactions, one involving the E. faecalis virulence factor ElrA and the human protein FHL2 was evidenced by yeast two-hybrid and biochemically confirmed. Further molecular characterizations allowed defining an FHL2-interacting domain (FID) of ElrA. Deletion of the FID led to an attenuated in vivo phenotype of the mutated strain clearly indicating that this interaction is likely to contribute to the multifactorial virulence of this opportunistic pathogen. Altogether, our results show that FHL2 is the first host cellular protein directly targeted by an E. faecalis virulence factor and that this interaction is involved in Enterococcus pathogenicity.
- Published
- 2017
- Full Text
- View/download PDF
32. Commensal gut bacteria modulate phosphorylation-dependent PPARγ transcriptional activity in human intestinal epithelial cells.
- Author
-
Nepelska M, de Wouters T, Jacouton E, Béguet-Crespel F, Lapaque N, Doré J, Arulampalam V, and Blottière HM
- Subjects
- Angiopoietin-Like Protein 4 metabolism, Bacteria, Anaerobic metabolism, Butyrates metabolism, Cell Culture Techniques, Cell Line, Culture Media, Conditioned, Humans, MAP Kinase Signaling System, Perilipin-2 metabolism, Phosphorylation, Propionates metabolism, Bacteria, Anaerobic growth & development, Epithelial Cells physiology, Gastrointestinal Microbiome, Gene Expression Regulation, Intestinal Mucosa physiology, PPAR gamma metabolism, Protein Processing, Post-Translational
- Abstract
In healthy subjects, the intestinal microbiota interacts with the host's epithelium, regulating gene expression to the benefit of both, host and microbiota. The underlying mechanisms remain poorly understood, however. Although many gut bacteria are not yet cultured, constantly growing culture collections have been established. We selected 57 representative commensal bacterial strains to study bacteria-host interactions, focusing on PPARγ, a key nuclear receptor in colonocytes linking metabolism and inflammation to the microbiota. Conditioned media (CM) were harvested from anaerobic cultures and assessed for their ability to modulate PPARγ using a reporter cell line. Activation of PPARγ transcriptional activity was linked to the presence of butyrate and propionate, two of the main metabolites of intestinal bacteria. Interestingly, some stimulatory CMs were devoid of these metabolites. A Prevotella and an Atopobium strain were chosen for further study, and shown to up-regulate two PPARγ-target genes, ANGPTL4 and ADRP. The molecular mechanisms of these activations involved the phosphorylation of PPARγ through ERK1/2. The responsible metabolites were shown to be heat sensitive but markedly diverged in size, emphasizing the diversity of bioactive compounds found in the intestine. Here we describe different mechanisms by which single intestinal bacteria can directly impact their host's health through transcriptional regulation.
- Published
- 2017
- Full Text
- View/download PDF
33. Energy balance and obesity: what are the main drivers?
- Author
-
Romieu I, Dossus L, Barquera S, Blottière HM, Franks PW, Gunter M, Hwalla N, Hursting SD, Leitzmann M, Margetts B, Nishida C, Potischman N, Seidell J, Stepien M, Wang Y, Westerterp K, Winichagoon P, Wiseman M, and Willett WC
- Subjects
- Beverages, Colon microbiology, Developing Countries statistics & numerical data, Energy Intake, Exercise, Feeding Behavior, Humans, Income, Malnutrition epidemiology, Microbiota physiology, Obesity genetics, Obesity microbiology, Weight Gain, Energy Metabolism, Obesity epidemiology
- Abstract
Purpose: The aim of this paper is to review the evidence of the association between energy balance and obesity., Methods: In December 2015, the International Agency for Research on Cancer (IARC), Lyon, France convened a Working Group of international experts to review the evidence regarding energy balance and obesity, with a focus on Low and Middle Income Countries (LMIC)., Results: The global epidemic of obesity and the double burden, in LMICs, of malnutrition (coexistence of undernutrition and overnutrition) are both related to poor quality diet and unbalanced energy intake. Dietary patterns consistent with a traditional Mediterranean diet and other measures of diet quality can contribute to long-term weight control. Limiting consumption of sugar-sweetened beverages has a particularly important role in weight control. Genetic factors alone cannot explain the global epidemic of obesity. However, genetic, epigenetic factors and the microbiota could influence individual responses to diet and physical activity., Conclusion: Energy intake that exceeds energy expenditure is the main driver of weight gain. The quality of the diet may exert its effect on energy balance through complex hormonal and neurological pathways that influence satiety and possibly through other mechanisms. The food environment, marketing of unhealthy foods and urbanization, and reduction in sedentary behaviors and physical activity play important roles. Most of the evidence comes from High Income Countries and more research is needed in LMICs.
- Published
- 2017
- Full Text
- View/download PDF
34. TLR ligands and butyrate increase Pyy expression through two distinct but inter-regulated pathways.
- Author
-
Larraufie P, Doré J, Lapaque N, and Blottière HM
- Subjects
- Cell Line, Glucagon-Like Peptide 1 biosynthesis, Humans, NF-kappa B metabolism, Butyrates metabolism, Enteroendocrine Cells metabolism, Gene Expression Regulation, Peptide YY metabolism, Toll-Like Receptors agonists
- Abstract
The intestinal epithelium is an active barrier separating the host from its microbiota. It senses microbial compounds through expression of a wide range of receptors including the Toll-like receptors (TLRs). TLRs have been shown to regulate epithelium permeability or secretion of defensin by Paneth cells. However, the expression and function of TLRs in enteroendocrine L-cells, a specific subtype of intestinal cells secreting PYY and GLP-1, have not yet been assessed. PYY and GLP-1 are implicated in regulation of gut motility, food intake and insulin secretion, and are of great interest regarding obesity and type 2 diabetes. Using a cellular model of human L-cells and a reporter system for NF-κB activation pathway, we reported functional expression of TLRs in these cells. Stimulation with specific TLR-agonists increased expression of Pyy but not Proglucagon in an NF-κB-dependent manner. Moreover, the effect of TLR stimulation was additive to butyrate, a product of bacterial fermentation, on Pyy expression. Additionally, butyrate also increased Tlr expression, including Tlr4, and the NF-κB response to TLR stimulation. Altogether, our results demonstrated a role of TLRs in the modulation of Pyy expression and the importance of butyrate, a product of bacterial fermentation in regulation of microbial TLR-dependent sensing., (© 2016 John Wiley & Sons Ltd.)
- Published
- 2017
- Full Text
- View/download PDF
35. A fibrolytic potential in the human ileum mucosal microbiota revealed by functional metagenomic.
- Author
-
Patrascu O, Béguet-Crespel F, Marinelli L, Le Chatelier E, Abraham AL, Leclerc M, Klopp C, Terrapon N, Henrissat B, Blottière HM, Doré J, and Béra-Maillet C
- Subjects
- Bacteroides metabolism, Carbohydrate Metabolism, Carboxymethylcellulose Sodium metabolism, Chromosome Mapping, Clostridiales metabolism, Feces microbiology, Humans, Metagenome, Metagenomics, Xylans metabolism, Dietary Fiber metabolism, Dietary Fiber microbiology, Gastrointestinal Microbiome, Ileum microbiology
- Abstract
The digestion of dietary fibers is a major function of the human intestinal microbiota. So far this function has been attributed to the microorganisms inhabiting the colon, and many studies have focused on this distal part of the gastrointestinal tract using easily accessible fecal material. However, microbial fermentations, supported by the presence of short-chain fatty acids, are suspected to occur in the upper small intestine, particularly in the ileum. Using a fosmid library from the human ileal mucosa, we screened 20,000 clones for their activities against carboxymethylcellulose and xylans chosen as models of the major plant cell wall (PCW) polysaccharides from dietary fibres. Eleven positive clones revealed a broad range of CAZyme encoding genes from Bacteroides and Clostridiales species, as well as Polysaccharide Utilization Loci (PULs). The functional glycoside hydrolase genes were identified, and oligosaccharide break-down products examined from different polysaccharides including mixed-linkage β-glucans. CAZymes and PULs were also examined for their prevalence in human gut microbiome. Several clusters of genes of low prevalence in fecal microbiome suggested they belong to unidentified strains rather specifically established upstream the colon, in the ileum. Thus, the ileal mucosa-associated microbiota encompasses the enzymatic potential for PCW polysaccharide degradation in the small intestine.
- Published
- 2017
- Full Text
- View/download PDF
36. The gut microbiota and obesity
- Author
-
Blottière HM, Romieu I, Dossus L, and Willett WC
- Published
- 2017
37. [Impact of newly developed metagenomic tools on our knowledge of the gut microbiota and its role in human health: diagnostic and therapeutic issues].
- Author
-
Blottière HM and Doré J
- Subjects
- Animals, Humans, Metagenomics trends, Dysbiosis diagnosis, Dysbiosis therapy, Gastrointestinal Microbiome physiology, Health, Metagenomics methods
- Abstract
Over the last years, our vision of the intestinal microbiota and its contribution to human physiology has been fully revisited, thanks to metagenomics. Next generation sequencing allowed a full characterization of the microbiome. Quantitative metagenomic permitted a deep understanding of the intestinal ecosystem, its structure, and potential dysbiosis in several human pathologies. The microbiome may be used as biomarker of disease or of risk to progress toward a disease. A good understanding of the mechanisms of interaction between gut microbiota and its host is needed; functional metagenomic is a useful tool to identify genes and metabolites able to interact with host's cells. Overall, the microbiome science that is developing opens new avenue for diagnosis, discovery of new drugs and potential therapeutic approaches. It is also a target for modulation by food, with potential impact on health., (© 2016 médecine/sciences – Inserm.)
- Published
- 2016
- Full Text
- View/download PDF
38. Lactobacillus rhamnosus CNCMI-4317 Modulates Fiaf/Angptl4 in Intestinal Epithelial Cells and Circulating Level in Mice.
- Author
-
Jacouton E, Mach N, Cadiou J, Lapaque N, Clément K, Doré J, van Hylckama Vlieg JE, Smokvina T, and Blottière HM
- Subjects
- Angiopoietin-Like Protein 4, Angiopoietins genetics, Angiopoietins metabolism, Animals, HT29 Cells, Humans, Lactobacillus physiology, Mice, Mice, Inbred C57BL, Epithelial Cells metabolism, Intestines cytology, Lacticaseibacillus rhamnosus physiology
- Abstract
Background and Objectives: Identification of new targets for metabolic diseases treatment or prevention is required. In this context, FIAF/ANGPTL4 appears as a crucial regulator of energy homeostasis. Lactobacilli are often considered to display beneficial effect for their hosts, acting on different regulatory pathways. The aim of the present work was to study the effect of several lactobacilli strains on Fiaf gene expression in human intestinal epithelial cells (IECs) and on mice tissues to decipher the underlying mechanisms., Subjects and Methods: Nineteen lactobacilli strains have been tested on HT-29 human intestinal epithelial cells for their ability to regulate Fiaf gene expression by RT-qPCR. In order to determine regulated pathways, we analysed the whole genome transcriptome of IECs. We then validated in vivo bacterial effects using C57BL/6 mono-colonized mice fed with normal chow., Results: We identified one strain (Lactobacillus rhamnosus CNCMI-4317) that modulated Fiaf expression in IECs. This regulation relied potentially on bacterial surface-exposed molecules and seemed to be PPAR-γ independent but PPAR-α dependent. Transcriptome functional analysis revealed that multiple pathways including cellular function and maintenance, lymphoid tissue structure and development, as well as lipid metabolism were regulated by this strain. The regulation of immune system and lipid and carbohydrate metabolism was also confirmed by overrepresentation of Gene Ontology terms analysis. In vivo, circulating FIAF protein was increased by the strain but this phenomenon was not correlated with modulation Fiaf expression in tissues (except a trend in distal small intestine)., Conclusion: We showed that Lactobacillus rhamnosus CNCMI-4317 induced Fiaf expression in human IECs, and increased circulating FIAF protein level in mice. Moreover, this effect was accompanied by transcriptome modulation of several pathways including immune response and metabolism in vitro.
- Published
- 2015
- Full Text
- View/download PDF
39. Commensal Streptococcus salivarius Modulates PPARγ Transcriptional Activity in Human Intestinal Epithelial Cells.
- Author
-
Couvigny B, de Wouters T, Kaci G, Jacouton E, Delorme C, Doré J, Renault P, Blottière HM, Guédon E, and Lapaque N
- Subjects
- Angiopoietin-Like Protein 4, Angiopoietins biosynthesis, Angiopoietins genetics, Caco-2 Cells, Cell Line, Tumor, Epithelial Cells cytology, Epithelial Cells metabolism, Fatty Acid-Binding Proteins biosynthesis, Fatty Acid-Binding Proteins genetics, Gene Expression Regulation, HT29 Cells, Humans, Immunity, Mucosal immunology, Intestinal Mucosa cytology, Intestinal Mucosa microbiology, Microbiota immunology, PPAR gamma genetics, Streptococcus immunology, Symbiosis, Intestinal Mucosa metabolism, PPAR gamma biosynthesis, Streptococcus metabolism, Transcription, Genetic genetics, Transcriptional Activation genetics
- Abstract
The impact of commensal bacteria in eukaryotic transcriptional regulation has increasingly been demonstrated over the last decades. A multitude of studies have shown direct effects of commensal bacteria from local transcriptional activity to systemic impact. The commensal bacterium Streptococcus salivarius is one of the early bacteria colonizing the oral and gut mucosal surfaces. It has been shown to down-regulate nuclear transcription factor (NF-кB) in human intestinal cells, a central regulator of the host mucosal immune system response to the microbiota. In order to evaluate its impact on a further important transcription factor shown to link metabolism and inflammation in the intestine, namely PPARγ (peroxisome proliferator-activated receptor), we used human intestinal epithelial cell-lines engineered to monitor PPARγ transcriptional activity in response to a wide range of S. salivarius strains. We demonstrated that different strains from this bacterial group share the property to inhibit PPARγ activation independently of the ligand used. First attempts to identify the nature of the active compounds showed that it is a low-molecular-weight, DNase-, proteases- and heat-resistant metabolite secreted by S. salivarius strains. Among PPARγ-targeted metabolic genes, I-FABP and Angptl4 expression levels were dramatically reduced in intestinal epithelial cells exposed to S. salivarius supernatant. Both gene products modulate lipid accumulation in cells and down-regulating their expression might consequently affect host health. Our study shows that species belonging to the salivarius group of streptococci impact both host inflammatory and metabolic regulation suggesting a possible role in the host homeostasis and health.
- Published
- 2015
- Full Text
- View/download PDF
40. Functional metagenomics to decipher food-microbe-host crosstalk.
- Author
-
Larraufie P, de Wouters T, Potocki-Veronese G, Blottière HM, and Doré J
- Subjects
- Humans, Food, Intestines microbiology, Metagenomics methods, Microbiota physiology
- Abstract
The recent developments of metagenomics permit an extremely high-resolution molecular scan of the intestinal microbiota giving new insights and opening perspectives for clinical applications. Beyond the unprecedented vision of the intestinal microbiota given by large-scale quantitative metagenomics studies, such as the EU MetaHIT project, functional metagenomics tools allow the exploration of fine interactions between food constituents, microbiota and host, leading to the identification of signals and intimate mechanisms of crosstalk, especially between bacteria and human cells. Cloning of large genome fragments, either from complex intestinal communities or from selected bacteria, allows the screening of these biological resources for bioactivity towards complex plant polymers or functional food such as prebiotics. This permitted identification of novel carbohydrate-active enzyme families involved in dietary fibre and host glycan breakdown, and highlighted unsuspected bacterial players at the top of the intestinal microbial food chain. Similarly, exposure of fractions from genomic and metagenomic clones onto human cells engineered with reporter systems to track modulation of immune response, cell proliferation or cell metabolism has allowed the identification of bioactive clones modulating key cell signalling pathways or the induction of specific genes. This opens the possibility to decipher mechanisms by which commensal bacteria or candidate probiotics can modulate the activity of cells in the intestinal epithelium or even in distal organs such as the liver, adipose tissue or the brain. Hence, in spite of our inability to culture many of the dominant microbes of the human intestine, functional metagenomics open a new window for the exploration of food-microbe-host crosstalk.
- Published
- 2015
- Full Text
- View/download PDF
41. The impact of ATRA on shaping human myeloid cell responses to epithelial cell-derived stimuli and on T-lymphocyte polarization.
- Author
-
Chatterjee A, Gogolak P, Blottière HM, and Rajnavölgyi É
- Subjects
- CD4-Positive T-Lymphocytes cytology, Caco-2 Cells, Cell Movement, Chemokines metabolism, Chemotaxis, Coculture Techniques, Dendritic Cells cytology, Flow Cytometry, Gene Expression Regulation, HCT116 Cells, HT29 Cells, Humans, Inflammation metabolism, Interleukin-17 metabolism, Interleukin-1beta metabolism, Lymphocyte Activation, Macrophages cytology, Monocytes cytology, Tumor Necrosis Factor-alpha metabolism, Epithelial Cells metabolism, Myeloid Cells cytology, Th17 Cells cytology, Tretinoin chemistry
- Abstract
Vitamin A plays an essential role in the maintenance of gut homeostasis but its interplay with chemokines has not been explored so far. Using an in vitro model system we studied the effects of human colonic epithelial cells (Caco2, HT-29, and HCT116) derived inflammatory stimuli on monocyte-derived dendritic cells and macrophages. Unstimulated Caco2 and HT-29 cells secreted CCL19, CCL21, and CCL22 chemokines, which could attract dendritic cells and macrophages and induced CCR7 receptor up-regulation by retinoic-acid resulting in dendritic cell migration. The chemokines Mk, CXCL16, and CXCL7 were secreted by all the 3 cell lines tested, and upon stimulation by IL-1β or TNF-α this effect was inhibited by ATRA but had no impact on CXCL1, CXCL8, and CCL20 secretion in response to IL-1β. In the presence of ATRA the supernatants of these cells induced CD103 expression on monocyte-derived dendritic cells and when conditioned by ATRA and cocultured with CD4(+) T-lymphocytes they reduced the proportion of Th17 T-cells. However, in the macrophage-T-cell cocultures the number of these effector T-cells was increased. Thus cytokine-activated colonic epithelial cells trigger the secretion of distinct combinations of chemokines depending on the proinflammatory stimulus and are controlled by retinoic acid, which also governs dendritic cell and macrophage responses.
- Published
- 2015
- Full Text
- View/download PDF
42. The laminin response in inflammatory bowel disease: protection or malignancy?
- Author
-
Spenlé C, Lefebvre O, Lacroute J, Méchine-Neuville A, Barreau F, Blottière HM, Duclos B, Arnold C, Hussenet T, Hemmerlé J, Gullberg D, Kedinger M, Sorokin L, Orend G, and Simon-Assmann P
- Subjects
- Animals, Caco-2 Cells, Cytokines metabolism, HCT116 Cells, HT29 Cells, Humans, Laminin genetics, Mice, Tumor Suppressor Protein p53 metabolism, Carcinoma metabolism, Colonic Neoplasms metabolism, Inflammatory Bowel Diseases metabolism, Laminin metabolism
- Abstract
Laminins (LM), basement membrane molecules and mediators of epithelial-stromal communication, are crucial in tissue homeostasis. Inflammatory Bowel Diseases (IBD) are multifactorial pathologies where the microenvironment and in particular LM play an important yet poorly understood role in tissue maintenance, and in cancer progression which represents an inherent risk of IBD. Here we showed first that in human IBD colonic samples and in murine colitis the LMα1 and LMα5 chains are specifically and ectopically overexpressed with a concomitant nuclear p53 accumulation. Linked to this observation, we provided a mechanism showing that p53 induces LMα1 expression at the promoter level by ChIP analysis and this was confirmed by knockdown in cell transfection experiments. To mimic the human disease, we induced colitis and colitis-associated cancer by chemical treatment (DSS) combined or not with a carcinogen (AOM) in transgenic mice overexpressing LMα1 or LMα5 specifically in the intestine. We demonstrated that high LMα1 or LMα5 expression decreased susceptibility towards experimentally DSS-induced colon inflammation as assessed by histological scoring and decrease of pro-inflammatory cytokines. Yet in a pro-oncogenic context, we showed that LM would favor tumorigenesis as revealed by enhanced tumor lesion formation in both LM transgenic mice. Altogether, our results showed that nuclear p53 and associated overexpression of LMα1 and LMα5 protect tissue from inflammation. But in a mutation setting, the same LM molecules favor progression of IBD into colitis-associated cancer. Our transgenic mice represent attractive new models to acquire knowledge about the paradoxical effect of LM that mediate either tissue reparation or cancer according to the microenvironment. In the early phases of IBD, reinforcing basement membrane stability/organization could be a promising therapeutic approach.
- Published
- 2014
- Full Text
- View/download PDF
43. A robust and adaptable high throughput screening method to study host-microbiota interactions in the human intestine.
- Author
-
de Wouters T, Ledue F, Nepelska M, Doré J, Blottière HM, and Lapaque N
- Subjects
- HT29 Cells, Host-Pathogen Interactions, Humans, Metagenome, High-Throughput Screening Assays methods, Intestines microbiology, Microbiota
- Abstract
The intestinal microbiota has many beneficial roles for its host. However, the precise mechanisms developed by the microbiota to influence the host intestinal cell responses are only partially known. The complexity of the ecosystem and our inability to culture most of these micro-organisms have led to the development of molecular approaches such as functional metagenomics, i.e. the heterologous expression of a metagenome in order to identify functions. This elegant strategy coupled to high throughput screening allowed to identify novel enzymes from different ecosystems where culture methods have not yet been adapted to isolate the candidate microorganisms. We have proposed to use this functional metagenomic approach in order to model the microbiota's interaction with the host by combining this heterologous expression with intestinal reporter cell lines. The addition of the cellular component to this functional metagenomic approach introduced a second important source of variability resulting in a novel challenge for high throughput screening. First attempts of high throughput screening with various reporter cell-lines showed a high distribution of the response and consequent difficulties to reproduce the response, impairing an easy and clear identification of confirmed hits. In this study, we developed a robust and reproducible methodology to combine these two biological systems for high throughput application. We optimized experimental setups and completed them by appropriate statistical analysis tools allowing the use this innovative approach in a high throughput manner and on a broad range of reporter assays. We herewith present a methodology allowing a high throughput screening combining two biological systems. Therefore ideal conditions for homogeneity, sensitivity and reproducibility of both metagenomic clones as well as reporter cell lines have been identified and validated. We believe that this innovative method will allow the identification of new bioactive microbial molecules and, subsequently, will promote understanding of host-microbiota interactions.
- Published
- 2014
- Full Text
- View/download PDF
44. Genome Sequence of "Candidatus Arthromitus" sp. Strain SFB-Mouse-NL, a Commensal Bacterium with a Key Role in Postnatal Maturation of Gut Immune Functions.
- Author
-
Bolotin A, de Wouters T, Schnupf P, Bouchier C, Loux V, Rhimi M, Jamet A, Dervyn R, Boudebbouze S, Blottière HM, Sorokin A, Snel J, Cerf-Bensussan N, Gaboriau-Routhiau V, van de Guchte M, and Maguin E
- Abstract
"Candidatus Arthromitus" sp. strain SFB-mouse-NL (SFB, segmented filamentous bacteria) is a commensal bacterium necessary for inducing the postnatal maturation of homeostatic innate and adaptive immune responses in the mouse gut. Here, we report the genome sequence of this bacterium, which sets it apart from earlier sequenced mouse SFB isolates., (Copyright © 2014 Bolotin et al.)
- Published
- 2014
- Full Text
- View/download PDF
45. Anti-inflammatory properties of Streptococcus salivarius, a commensal bacterium of the oral cavity and digestive tract.
- Author
-
Kaci G, Goudercourt D, Dennin V, Pot B, Doré J, Ehrlich SD, Renault P, Blottière HM, Daniel C, and Delorme C
- Subjects
- Animals, Colitis immunology, Colitis pathology, Disease Models, Animal, Epithelial Cells immunology, Epithelial Cells microbiology, Humans, Leukocytes, Mononuclear immunology, Leukocytes, Mononuclear microbiology, Mice, NF-kappa B antagonists & inhibitors, NF-kappa B immunology, Anti-Inflammatory Agents metabolism, Gastrointestinal Tract microbiology, Mouth microbiology, Streptococcus immunology, Streptococcus physiology, Symbiosis
- Abstract
Streptococcus salivarius is one of the first colonizers of the human oral cavity and gut after birth and therefore may contribute to the establishment of immune homeostasis and regulation of host inflammatory responses. The anti-inflammatory potential of S. salivarius was first evaluated in vitro on human intestinal epithelial cells and human peripheral blood mononuclear cells. We show that live S. salivarius strains inhibited in vitro the activation of the NF-κB pathway on intestinal epithelial cells. We also demonstrate that the live S. salivarius JIM8772 strain significantly inhibited inflammation in severe and moderate colitis mouse models. These in vitro and in vivo anti-inflammatory properties were not found with heat-killed S. salivarius, suggesting a protective response exclusively with metabolically active bacteria.
- Published
- 2014
- Full Text
- View/download PDF
46. High-throughput system for the presentation of secreted and surface-exposed proteins from Gram-positive bacteria in functional metagenomics studies.
- Author
-
Dobrijevic D, Di Liberto G, Tanaka K, de Wouters T, Dervyn R, Boudebbouze S, Binesse J, Blottière HM, Jamet A, Maguin E, and van de Guchte M
- Subjects
- Bacillus subtilis metabolism, Bacterial Proteins genetics, Bacterial Proteins metabolism, Escherichia coli genetics, Flagellin biosynthesis, Flagellin immunology, Flagellin metabolism, Gene Expression, HT29 Cells, Humans, Immunity, Cellular, Membrane Proteins genetics, Metagenomics, Plasmids genetics, Recombinant Proteins biosynthesis, Recombinant Proteins genetics, Recombinant Proteins metabolism, Staphylococcus aureus genetics, Bacillus subtilis genetics, Bacterial Proteins biosynthesis, Cloning, Molecular methods, Membrane Proteins biosynthesis
- Abstract
Complex microbial ecosystems are increasingly studied through the use of metagenomics approaches. Overwhelming amounts of DNA sequence data are generated to describe the ecosystems, and allow to search for correlations between gene occurrence and clinical (e.g. in studies of the gut microbiota), physico-chemical (e.g. in studies of soil or water environments), or other parameters. Observed correlations can then be used to formulate hypotheses concerning microbial gene functions in relation to the ecosystem studied. In this context, functional metagenomics studies aim to validate these hypotheses and to explore the mechanisms involved. One possible approach is to PCR amplify or chemically synthesize genes of interest and to express them in a suitable host in order to study their function. For bacterial genes, Escherichia coli is often used as the expression host but, depending on the origin and nature of the genes of interest and the test system used to evaluate their putative function, other expression systems may be preferable. In this study, we developed a system to evaluate the role of secreted and surface-exposed proteins from Gram-positive bacteria in the human gut microbiota in immune modulation. We chose to use a Gram-positive host bacterium, Bacillus subtilis, and modified it to provide an expression background that behaves neutral in a cell-based immune modulation assay, in vitro. We also adapted an E. coli-B. subtilis shuttle expression vector for use with the Gateway high-throughput cloning system. Finally, we demonstrate the functionality of this host-vector system through the cloning and expression of a flagellin-coding sequence, and show that the expression-clone elicits an inflammatory response in a human intestinal epithelial cell line. The expression host can easily be adapted to assure neutrality in other assay systems, allowing the use of the presented presentation system in functional metagenomics of the gut and other ecosystems.
- Published
- 2013
- Full Text
- View/download PDF
47. ANGPTL4 expression induced by butyrate and rosiglitazone in human intestinal epithelial cells utilizes independent pathways.
- Author
-
Korecka A, de Wouters T, Cultrone A, Lapaque N, Pettersson S, Doré J, Blottière HM, and Arulampalam V
- Subjects
- Angiopoietin-Like Protein 4, Angiopoietins genetics, Animals, Caco-2 Cells, Clostridium tyrobutyricum, Enterocytes metabolism, Germ-Free Life, HCT116 Cells, HT29 Cells, Humans, Intestinal Mucosa microbiology, Metagenome, Mice, Mice, Inbred C57BL, PPAR gamma agonists, PPAR gamma metabolism, Response Elements, Rosiglitazone, Transcription Initiation Site, Angiopoietins metabolism, Butyrates pharmacology, Hypoglycemic Agents pharmacology, Intestinal Mucosa metabolism, Thiazolidinediones pharmacology, Transcription, Genetic drug effects
- Abstract
Short-chain fatty acids (SCFAs), such as butyrate and propionate, are metabolic products of carbohydrate fermentation by the microbiota and constitute the main source of energy for host colonocytes. SCFAs are also important for gastrointestinal health, immunity, and host metabolism. Intestinally produced angiopoietin-like protein 4 (ANGPTL4) is a secreted protein with metabolism-altering properties and may offer a route by which microbiota can regulate host metabolism. Peroxisome proliferator-activated receptor (PPAR)-γ has previously been shown to be involved in microbiota-induced expression of intestinal ANGPTL4, but the role of bacterial metabolites in this process has remained elusive. Here, we show that the SCFA butyrate regulates intestinal ANGPTL4 expression in a PPAR-γ-independent manner. Although PPAR-γ is not required for butyrate-driven intestinal ANGPTL4 expression, costimulating with PPAR-γ ligands and SCFAs leads to additive increases in ANGPTL4 levels. We suggest that PPAR-γ and butyrate rely on two separate regulatory sites, a PPAR-responsive element downstream the transcription start site and a butyrate-responsive element(s) within the promoter region, 0.5 kb upstream of the transcription start site. Furthermore, butyrate gavage and colonization with Clostridium tyrobutyricum, a SCFA producer, can independently induce expression of intestinal ANGPTL4 in germ-free mice. Thus, oral administration of SCFA or use of SCFA-producing bacteria may be additional routes to maintain intestinal ANGPTL4 levels for preventive nutrition or therapeutic purposes.
- Published
- 2013
- Full Text
- View/download PDF
48. Human intestinal metagenomics: state of the art and future.
- Author
-
Blottière HM, de Vos WM, Ehrlich SD, and Doré J
- Subjects
- Humans, Gastrointestinal Tract microbiology, Metagenomics methods, Microbiota
- Abstract
Over the last few years our understanding of human biology has undergone profound transformation. The key role of the 'world inside us', namely the gut microbiota, once considered a forgotten organ, has been revealed, with strong impact on our health and well-being. The present review highlights the most important recent findings on the role of gut microbiota and its impact on the host and raises crucial questions to be considered in future studies., (Copyright © 2013 Elsevier Ltd. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
49. The NF-κB binding site located in the proximal region of the TSLP promoter is critical for TSLP modulation in human intestinal epithelial cells.
- Author
-
Cultrone A, de Wouters T, Lakhdari O, Kelly D, Mulder I, Logan E, Lapaque N, Doré J, and Blottière HM
- Subjects
- Base Sequence, Binding Sites, Cell Line, Cytokines metabolism, Gene Expression Regulation drug effects, Gene Order, Humans, Interleukin-1 pharmacology, Mitogen-Activated Protein Kinases metabolism, Neurofibromin 1 metabolism, Neurofibromin 2 metabolism, Protein Binding, Transcription Factor AP-1 metabolism, Thymic Stromal Lymphopoietin, Cytokines genetics, Epithelial Cells metabolism, Intestinal Mucosa metabolism, NF-kappa B metabolism, Promoter Regions, Genetic
- Abstract
Thymic stromal lymphopoietin (TSLP) is constitutively secreted by intestinal epithelial cells. It regulates gut DCs, therefore, contributing to the maintenance of immune tolerance. In the present report, we describe the regulation of TSLP expression in intestinal epithelial cells and characterize the role of several NF-κB binding sites present on the TSLP promoter. TSLP expression can be stimulated by different compounds through activation of p38, protein kinase A, and finally the NF-κB pathway. We describe a new NF-κB binding element located at position -0.37 kb of the promoter that is crucial for the NF-κB-dependent regulation of TSLP. We showed that mutation of this proximal NF-κB site abrogates the IL-1β-mediated transcriptional activation of human TSLP in several epithelial cell lines. We also demonstrated that both p65 and p50 subunits are able to bind this new NF-κB binding site. The present work provides new insight into epithelial cell-specific TSLP regulation., (© 2013 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim.)
- Published
- 2013
- Full Text
- View/download PDF
50. A metagenomic insight into our gut's microbiome.
- Author
-
Lepage P, Leclerc MC, Joossens M, Mondot S, Blottière HM, Raes J, Ehrlich D, and Doré J
- Subjects
- Bacteriophages, Humans, Inflammatory Bowel Diseases microbiology, Metagenome physiology, Intestines microbiology, Metagenome genetics, Metagenomics methods
- Abstract
Advances in sequencing technology and the development of metagenomic and bioinformatics methods have opened up new ways to investigate the 10(14) microorganisms inhabiting the human gut. The gene composition of human gut microbiome in a large and deeply sequenced cohort highlighted an overall non-redundant genome size 150 times larger than the human genome. The in silico predictions based on metagenomic sequencing are now actively followed, compared and challenged using additional 'omics' technologies. Interactions between the microbiota and its host are of key interest in several pathologies and applying meta-omics to describe the human gut microbiome will give a better understanding of this crucial crosstalk at mucosal interfaces. Adding to the growing appreciation of the importance of the microbiome is the discovery that numerous phages, that is, viruses of prokaryotes infecting bacteria (bacteriophages) or archaea with a high host specificity, inhabit the human gut and impact microbial activity. In addition, gene exchanges within the gut microbiota have proved to be more frequent than anticipated. Taken together, these innovative exploratory technologies are expected to unravel new information networks critical for gut homeostasis and human health. Among the challenges faced, the in vivo validation of these networks, together with their integration into the prediction and prognosis of disease, may require further working hypothesis and collaborative efforts.
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.