101
|
Rabiei N, Ahmadi Badi S, Ettehad Marvasti F, Nejad Sattari T, Vaziri F, Siadat SD. Induction effects of Faecalibacterium prausnitzii and its extracellular vesicles on toll-like receptor signaling pathway gene expression and cytokine level in human intestinal epithelial cells. Cytokine 2019; 121:154718. [PMID: 31153056 DOI: 10.1016/j.cyto.2019.05.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 04/15/2019] [Accepted: 05/08/2019] [Indexed: 02/08/2023]
Abstract
A single layer of epithelial cells creates an interface between the host and microorganisms colonizing the gastrointestinal tract. In a healthy intestine, commensal bacteria and their metabolites can interact with epithelial cells as they are identified by Toll-like receptors (TLRs); This interaction results in homeostasis and immune responses. The present study aimed at evaluating Faecalibacterium prausnitzii- and extracellular vesicles (EVs)-induced expression of involved genes in TLRs signaling pathway and cytokines production in Caco-2 cell line. In this study, Caco-2 cell line was treated with F. prausitzii and its EVs. Using the protein levels of 12 cytokines were also evaluated by ELISA assay. F. prausnitzii induced upregulation in FOS, JUN, TNF-α, NFKB1, TLR3, IKBKB and CD86 genes. Furthermore, stimulation of Caco-2 cells with EVs derived from F. prausnitzii induced upregulation of CXCL8, CCL2, FOS, MAP2K4, TLR7, TLR3, IRF1, NFKBIA and TNF-α genes. Based on ELISA assay, Caco-2 cells treated with F. prausnitzii and its EVs showed a significant increase in TNF-α, IL-4, IL-8, and IL-10 expression and significant decreased in IL-1, IL-2, IL-6, IL-12, IL-17a, IFN-γ compared to the control group (P < 0.05). In conclusion, EVs derived from F. prausnitzii showed greater efficacy in decreasing the inflammatory cytokines and increasing the anti-inflammatory cytokines, compared to F. prausnitzii. Our findings can be used as a theoretical model for EVs application in the potential treatment of inflammation.
Collapse
Affiliation(s)
- Nasrin Rabiei
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Sara Ahmadi Badi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran; Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran
| | | | - Taher Nejad Sattari
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Farzam Vaziri
- Mycobacteriology and Pulmonary Research Department, Pasteur Institute of Iran, Tehran, Iran; Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran
| | - Seyed Davar Siadat
- Mycobacteriology and Pulmonary Research Department, Pasteur Institute of Iran, Tehran, Iran; Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
102
|
Almeida D, Machado D, Andrade JC, Mendo S, Gomes AM, Freitas AC. Evolving trends in next-generation probiotics: a 5W1H perspective. Crit Rev Food Sci Nutr 2019; 60:1783-1796. [PMID: 31062600 DOI: 10.1080/10408398.2019.1599812] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
In recent years, scientific community has been gathering increasingly more insight on the dynamics that are at play in metabolic and inflammatory disorders. These rapidly growing conditions are reaching epidemic proportions, bringing clinicians and researcher's new challenges. The specific roles and modulating properties that beneficial/probiotic bacteria hold in the context of the gut ecosystem seem to be key to avert these inflammatory and diet-related disorders. Faecalibacterium prausnitzii, Akkermansia muciniphila and Eubacterium hallii have been identified as candidates for next generation probiotics (NGPs) with exciting potential for the prevention and treatment of such of dysbiosis-associated diseases. The challenges of these non-conventional native gut bacteria lie mainly on their extreme sensitivity to O2 traces. If these strains are to be used successfully in food, supplements or drugs they need to be stable and active in humans. In the present review, we present an overall perspective of the most updated scientific literature on the newly called NGPs through the 5W1H (What, Why, Who, Where, When, and How) method, an innovative and attractive problem-solving approach that provides the reader an effective understanding of the issue at hand.
Collapse
Affiliation(s)
- Diana Almeida
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Porto, Portugal.,Department of Biology and CESAM, University of Aveiro, Aveiro, Portugal
| | - Daniela Machado
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Porto, Portugal
| | - José Carlos Andrade
- CESPU, Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde, Gandra PRD, Portugal
| | - Sónia Mendo
- Department of Biology and CESAM, University of Aveiro, Aveiro, Portugal
| | - Ana Maria Gomes
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Porto, Portugal
| | - Ana Cristina Freitas
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Porto, Portugal
| |
Collapse
|
103
|
The potential probiotic Lactobacillus rhamnosus CNCM I-3690 strain protects the intestinal barrier by stimulating both mucus production and cytoprotective response. Sci Rep 2019; 9:5398. [PMID: 30931953 PMCID: PMC6443702 DOI: 10.1038/s41598-019-41738-5] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 03/11/2019] [Indexed: 12/14/2022] Open
Abstract
The gut barrier plays an important role in human health. When barrier function is impaired, altered permeability and barrier dysfunction can occur, leading to inflammatory bowel diseases, irritable bowel syndrome or obesity. Several bacteria, including pathogens and commensals, have been found to directly or indirectly modulate intestinal barrier function. The use of probiotic strains could be an important landmark in the management of gut dysfunction with a clear impact on the general population. Previously, we found that Lactobacillus rhamnosus CNCM I-3690 can protect intestinal barrier functions in mice inflammation model. Here, we investigated its mechanism of action. Our results show that CNCM I-3690 can (i) physically maintain modulated goblet cells and the mucus layer and (ii) counteract changes in local and systemic lymphocytes. Furthermore, mice colonic transcriptome analysis revealed that CNCM I-3690 enhances the expression of genes related to healthy gut permeability: motility and absorption, cell proliferation; and protective functions by inhibiting endogenous proteases. Finally, SpaFED pili are clearly important effectors since an L. rhamnosus ΔspaF mutant failed to provide the same benefits as the wild type strain. Taken together, our data suggest that CNCM I-3690 restores impaired intestinal barrier functions via anti-inflammatory and cytoprotective responses.
Collapse
|
104
|
Peters VBM, van de Steeg E, van Bilsen J, Meijerink M. Mechanisms and immunomodulatory properties of pre- and probiotics. Benef Microbes 2019; 10:225-236. [PMID: 30827150 DOI: 10.3920/bm2018.0066] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The human body is exposed to many xenobiotic, potentially harmful compounds. The intestinal immune system is crucial in protecting the human body from these substances. Moreover, many microorganisms, residing in the gastrointestinal tract, play an important role in modulating immune responses. Pre- and probiotics may have beneficial effects on the microbial composition and activity within the human gut, subsequently affecting the immune system. Prebiotics can exert their effects via different mechanisms, like selectively stimulating the growth of bacteria by providing substrates or via direct immune stimulation. Probiotics may have beneficial health effects via competition with pathogens for substrates and binding intestinal sites, bioconversions of for example sugars into fermentation products with inhibitory properties, production of growth substrates like vitamins for the host, direct antagonism of pathogens via antimicrobial peptide production, reduction of inflammation and stimulation of immune cells. This review focuses on the different mechanisms via which the pre- and probiotics exert their beneficial effects on the host, addressing their immunomodulatory properties in particular.
Collapse
Affiliation(s)
- V B M Peters
- 1 TNO Zeist, Department Microbiology and Systems Biology, Utrechtseweg 48, 3704 HE Zeist, the Netherlands.,2 University College London, Division of Medicine, Gower Street, WC1E 6BT London, United Kingdom
| | - E van de Steeg
- 1 TNO Zeist, Department Microbiology and Systems Biology, Utrechtseweg 48, 3704 HE Zeist, the Netherlands
| | - J van Bilsen
- 3 TNO Zeist, Department Risk Analysis for Products in Development (RAPID), Utrechtseweg 48, 3704 HE Zeist, the Netherlands
| | - M Meijerink
- 3 TNO Zeist, Department Risk Analysis for Products in Development (RAPID), Utrechtseweg 48, 3704 HE Zeist, the Netherlands
| |
Collapse
|
105
|
Ameliorative effect of salidroside from Rhodiola Rosea L. on the gut microbiota subject to furan-induced liver injury in a mouse model. Food Chem Toxicol 2019; 125:333-340. [DOI: 10.1016/j.fct.2019.01.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Revised: 01/08/2019] [Accepted: 01/10/2019] [Indexed: 01/06/2023]
|
106
|
Renouf MJ, Cho YH, McPhee JB. Emergent Behavior of IBD-Associated Escherichia coli During Disease. Inflamm Bowel Dis 2019; 25:33-44. [PMID: 30321333 DOI: 10.1093/ibd/izy312] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Indexed: 12/12/2022]
Abstract
Inflammatory bowel diseases are becoming increasingly common throughout the world, both in developed countries and increasingly in rapidly developing countries. Multiple lines of evidence point to a role for the microbial composition of the gastrointestinal tract in the etiology of IBD, but to date, attempts to define a specific microbial cause for IBD have proved unsuccessful. Microbial 16S rRNA profiling shows that IBD patients have elevated levels of Enterobacteriaceae, in particular Escherichia coli, and reduced levels of Faecalibacterium prausnitzii. The observed E. coli have been assigned to a specific pathovar, adherent-invasive E. coli (AIEC). Adherent-invasive E. coli are a genomically heterogenous group, and whereas many groups have attempted to identify specific genetic markers that differentiate AIEC from non-AIEC strains, very few concrete genetic associations have been uncovered. Here, we highlight the advantages of applying a phenotyping approach to the study of these organisms, rather than solely depending on a sequencing or genomic-based screening strategy because virulence-associated phenotypes exhibit behaviors of emergent systems. In this respect, attempts at genetic reductionism are prone to failure because there are numerous metabolic, regulatory or genetic paths that can underlie these virulence-associated behaviors. Here, we review these IBD-associated phenotypes in E. coli and make recommendations for experimental approaches to advance our understanding of IBD-associated bacteria more generally. With advances in high-throughput screening and nongenetically based metabolomic characterization of IBD-associated bacteria, we anticipate a fuller understanding of how altered microbial communities contribute to the development of IBD.
Collapse
Affiliation(s)
| | - Youn Hee Cho
- Department of Chemistry and Biology, Ryerson University, Toronto ON, Canada
| | - Joseph B McPhee
- Department of Chemistry and Biology, Ryerson University, Toronto ON, Canada
| |
Collapse
|
107
|
Rolig AS, Sweeney EG, Kaye LE, DeSantis MD, Perkins A, Banse AV, Hamilton MK, Guillemin K. A bacterial immunomodulatory protein with lipocalin-like domains facilitates host-bacteria mutualism in larval zebrafish. eLife 2018; 7:e37172. [PMID: 30398151 PMCID: PMC6219842 DOI: 10.7554/elife.37172] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Accepted: 10/04/2018] [Indexed: 02/06/2023] Open
Abstract
Stable mutualism between a host and its resident bacteria requires a moderated immune response to control bacterial population size without eliciting excessive inflammation that could harm both partners. Little is known about the specific molecular mechanisms utilized by bacterial mutualists to temper their hosts' responses and protect themselves from aggressive immune attack. Using a gnotobiotic larval zebrafish model, we identified an Aeromonas secreted immunomodulatory protein, AimA. AimA is required during colonization to prevent intestinal inflammation that simultaneously compromises both bacterial and host survival. Administration of exogenous AimA prevents excessive intestinal neutrophil accumulation and protects against septic shock in models of both bacterially and chemically induced intestinal inflammation. We determined the molecular structure of AimA, which revealed two related calycin-like domains with structural similarity to the mammalian immune modulatory protein, lipocalin-2. As a secreted bacterial protein required by both partners for optimal fitness, AimA is an exemplar bacterial mutualism factor.
Collapse
Affiliation(s)
- Annah S Rolig
- Institute of Molecular BiologyUniversity of OregonEugeneUnited States
| | | | - Lila E Kaye
- Institute of Molecular BiologyUniversity of OregonEugeneUnited States
| | | | - Arden Perkins
- Institute of Molecular BiologyUniversity of OregonEugeneUnited States
| | - Allison V Banse
- Institute of Molecular BiologyUniversity of OregonEugeneUnited States
| | | | - Karen Guillemin
- Institute of Molecular BiologyUniversity of OregonEugeneUnited States
- Humans and the Microbiome ProgramCanadian Institute for Advanced ResearchTorontoCanada
| |
Collapse
|
108
|
Salaga M, Binienda A, Draczkowski P, Kosson P, Kordek R, Jozwiak K, Fichna J. Novel peptide inhibitor of dipeptidyl peptidase IV (Tyr-Pro-D-Ala-NH 2) with anti-inflammatory activity in the mouse models of colitis. Peptides 2018; 108:34-45. [PMID: 30179653 DOI: 10.1016/j.peptides.2018.08.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 08/03/2018] [Accepted: 08/24/2018] [Indexed: 12/19/2022]
Abstract
Protease inhibition has become a new possible approach in the inflammatory bowel disease (IBD) therapy. A serine exopeptidase, dipeptidyl peptidase IV (DPP IV) is responsible for inactivation of incretin hormone, glucagon-like peptide 2 (GLP-2), a potent stimulator of intestinal epithelium regeneration and growth. Recently we showed that the novel peptide analog of endomorphin-2, EMDB-1 (Tyr-Pro-D-ClPhe-Phe-NH2) is a potent blocker of DPP IV and exhibits an anti-inflammatory activity in vivo. The aim of this study was to design, synthesize and characterize the therapeutic activity and mechanism of action of a series of novel EMDB-1 analogs. The inhibitory potential of all peptides was evaluated using the fluorometric screening assay employing Gly-Pro-Aminomethylcoumarin (AMC) to measure DPP IV activity. Consequently, one compound, namely DI-1 was selected and its therapeutic activity evaluated using mouse models of experimental colitis (induced by TNBS and DSS). Macro- and microscopic score, ulcer score, colonic wall thickness as well as myeloperoxidase activity were measured. We showed that DI-1 blocks DPP IV in vitro (IC50 = 0.76 ± 0.04 nM) and attenuates acute, semichronic and relapsing TNBS- as well as DSS-induced colitis in mice after topical administration. Its anti-inflammatory action is associated with the increase of colonic GLP-2 but not GLP2 receptor or DPP IV expression. Our results validate DPP IV as a pharmacological target for the anti-IBD drugs and its inhibitors, such as DI-1, have the potential to become valuable anti-inflammatory therapeutics.
Collapse
Affiliation(s)
- M Salaga
- Department of Biochemistry, Department of Pathology, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - A Binienda
- Department of Biochemistry, Department of Pathology, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - P Draczkowski
- Department of Biopharmacy, Medical University of Lublin, Poland
| | - P Kosson
- Mossakowski Medical Research Centre Polish Academy of Sciences, Warsaw, Poland
| | - R Kordek
- Department of Biochemistry, Department of Pathology, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - K Jozwiak
- Department of Biopharmacy, Medical University of Lublin, Poland
| | - J Fichna
- Department of Biochemistry, Department of Pathology, Faculty of Medicine, Medical University of Lodz, Lodz, Poland.
| |
Collapse
|
109
|
Godefroy E, Alameddine J, Montassier E, Mathé J, Desfrançois-Noël J, Marec N, Bossard C, Jarry A, Bridonneau C, Le Roy A, Sarrabayrouse G, Kerdreux E, Bourreille A, Sokol H, Jotereau F, Altare F. Expression of CCR6 and CXCR6 by Gut-Derived CD4 +/CD8α + T-Regulatory Cells, Which Are Decreased in Blood Samples From Patients With Inflammatory Bowel Diseases. Gastroenterology 2018; 155:1205-1217. [PMID: 29981781 DOI: 10.1053/j.gastro.2018.06.078] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 06/27/2018] [Accepted: 06/30/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Faecalibacterium prausnitzii, a member of the Clostridium IV group of the Firmicutes phylum that is abundant in the intestinal microbiota, has anti-inflammatory effects. The relative level of F prausnitzii is decreased in fecal samples from patients with inflammatory bowel diseases (IBDs) compared with healthy individuals. Reduced F prausnitzii was correlated with relapse of Crohn's disease after surgery. We identified, in human colonic mucosa and blood, a population of T regulatory type 1-like T regulatory (TREG) cells that express CD4 and CD8α (DP8α T cells) and are specific for F prausnitzii. We aimed to determine whether they are altered in patients with IBD. METHODS We isolated DP8α T cells from human colon lamina propria and blood samples and used flow cytometry to detect markers of cells that are of colon origin. We quantified DP8α cells that express colon-specific markers in blood samples from 106 patients with IBD, 12 patients with infectious colitis, and 35 healthy donors (controls). We identified cells that respond to F prausnitzii. Cells were stimulated with anti-CD3, and their production of interleukin 10 was measured by enzyme-linked immunosorbent assay. We compared the frequency and reactivity of cells from patients vs controls using the 2-sided Student t test or 1-way analysis of variance. RESULTS Circulating DP8α T cells that proliferate in response to F prausnitzii express the C-C motif chemokine receptor 6 (CCR6) and C-X-C motif chemokine receptor 6 (CXCR6). These cells also have features of TREG cells, including production of IL-10 and inhibition of T-cell proliferation via CD39 activity. The proportion of circulating CCR6+/CXCR6+ DP8α T cells was significantly reduced (P < .0001) within the total population of CD3+ T cells from patients with IBD compared with patients with infectious colitis or controls. A threshold of <7.875 CCR6+/CXCR6+ DP8α T cells/10,000 CD3+ cells discriminated patients with IBD from those with infectious colitis with 100% specificity and 72.2% sensitivity. CONCLUSIONS We identified a population of gut-derived TREG cells that are reduced in blood samples from patients with IBD compared with patients with infectious colitis or controls. These cells should be studied further to determine the mechanisms of this reduction and how it might contribute to the pathogenesis of IBD and their prognostic or diagnostic value.
Collapse
Affiliation(s)
| | - Joudy Alameddine
- CRCINA, INSERM, University of Nantes, University of Angers, Nantes, France
| | - Emmanuel Montassier
- MiHAR Lab, Institut de Recherche en Santé 2, Université de Nantes, Nantes, France; Emergency Department, Centre Hospitalier Universitaire de Nantes, Nantes, France
| | - Justine Mathé
- CRCINA, INSERM, University of Nantes, University of Angers, Nantes, France
| | | | | | - Céline Bossard
- INSERM U1232, IRS-UN, Nantes, France; Pathology Department, CHU Nantes, Nantes, France
| | | | - Chantal Bridonneau
- Commensal and Probiotic-Host Interactions Laboratory, INRA, Jouy-en-Josas, France
| | - Amandine Le Roy
- CRCINA, INSERM, University of Nantes, University of Angers, Nantes, France
| | | | - Elise Kerdreux
- CIC, INSERM 1413, CHU Nantes, Hôpital Hôtel-Dieu, Nantes, France; Institut des Maladies de l'Appareil Digestif, CHU Nantes, Hôpital Hôtel-Dieu, Nantes, France
| | - Arnaud Bourreille
- CIC, INSERM 1413, CHU Nantes, Hôpital Hôtel-Dieu, Nantes, France; Institut des Maladies de l'Appareil Digestif, CHU Nantes, Hôpital Hôtel-Dieu, Nantes, France; INSERM, UMR1235, Nantes, France; Université Nantes, Nantes, France
| | - Harry Sokol
- Commensal and Probiotic-Host Interactions Laboratory, INRA, Jouy-en-Josas, France; Sorbonne University-UPMC Université Paris 06, Ecole Normale Supérieure, CNRS, INSERM, AP-HP, Laboratoires des Biomolécules, Paris, France; Department of Gastroenterology, Saint Antoine Hospital, AP-HP, Paris, France
| | - Francine Jotereau
- CRCINA, INSERM, University of Nantes, University of Angers, Nantes, France.
| | - Frédéric Altare
- CRCINA, INSERM, University of Nantes, University of Angers, Nantes, France.
| |
Collapse
|
110
|
Lopez-Siles M, Enrich-Capó N, Aldeguer X, Sabat-Mir M, Duncan SH, Garcia-Gil LJ, Martinez-Medina M. Alterations in the Abundance and Co-occurrence of Akkermansia muciniphila and Faecalibacterium prausnitzii in the Colonic Mucosa of Inflammatory Bowel Disease Subjects. Front Cell Infect Microbiol 2018; 8:281. [PMID: 30245977 PMCID: PMC6137959 DOI: 10.3389/fcimb.2018.00281] [Citation(s) in RCA: 118] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 07/25/2018] [Indexed: 12/21/2022] Open
Abstract
Akkermansia muciniphila and Faecalibacterium prausnitzii, cohabitants in the intestinal mucosa, are considered members of a healthy microbiota and reduction of both species occurs in several intestinal disorders, including inflammatory bowel disease. Little is known however about a possible link between the reduction in quantity of these species, and in which circumstances this may occur. This study aims to determine the abundances and co-occurrence of the two species in order to elucidate conditions that may compromise their presence in the gut. Loads of A. muciniphila, total F. prausnitzii and its two phylogroup (16S rRNA gene copies) were determined by quantitative polymerase chain reaction in colonic biopsies from 17 healthy controls (H), 23 patients with ulcerative colitis (UC), 31 patients with Crohn's disease (CD), 3 with irritable bowel syndrome (IBS) and 3 with colorectal cancer (CRC). Data were normalized to total bacterial 16S rRNA gene copies in the same sample. Prevalence, relative abundances and correlation analyses were performed according to type of disease and considering relevant clinical characteristics of patients such as IBD location, age of disease onset, CD behavior, current medication and activity status. Co-occurrence of both species was found in 29% of H, 65% of UC and 29% of CD. Lower levels of total F. prausnitzii and phylogroups were found in subjects with CD, compared with H subjects (P ≤ 0.044). In contrast, no differences were found with the regard to A. muciniphila abundance across different disease states, but CD patients with disease onset below 16 years of age featured a marked depletion of this species. In CD patients, correlation between A. muciniphila and total F. prausnitzii (ρ = 0.362, P = 0.045) was observed, and particularly in those with non-stricturing, non-penetrating disease behavior and under moderate immunosuppressants therapy. Altogether, this study revealed that co-occurrence of both species differs between disease status. In addition, IBD patients featured a reduction of F. prausnitzii but similar loads of A. muciniphila when compared to H subjects, with the exception of those with early onset CD. Depletion of A. muciniphila in this subgroup of subjects suggests that it could be a potential biomarker to assist in pediatric CD diagnosis.
Collapse
Affiliation(s)
- Mireia Lopez-Siles
- Laboratory of Molecular Microbiology, Biology Department, Universitat de Girona, Girona, Spain
| | - Núria Enrich-Capó
- Laboratory of Molecular Microbiology, Biology Department, Universitat de Girona, Girona, Spain
| | - Xavier Aldeguer
- Department of Gastroenterology, Hospital Dr. Josep Trueta, Girona, Spain
| | - Miriam Sabat-Mir
- Department of Gastroenterology, Hospital Santa Caterina, Girona, Spain
| | - Sylvia H Duncan
- Microbiology Group, Rowett Institute of Nutrition and Health, Aberdeen, United Kingdom
| | - L Jesús Garcia-Gil
- Laboratory of Molecular Microbiology, Biology Department, Universitat de Girona, Girona, Spain
| | | |
Collapse
|
111
|
Broom LJ. Gut barrier function: Effects of (antibiotic) growth promoters on key barrier components and associations with growth performance. Poult Sci 2018; 97:1572-1578. [PMID: 29462405 DOI: 10.3382/ps/pey021] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 01/10/2018] [Indexed: 01/06/2023] Open
Abstract
The gut barrier, comprising the microbiota and their products, mucus layers, host-derived antimicrobial compounds [e.g., host defense peptides (HDP), IgA], epithelium, and underlying immune tissues, performs the essential function of preventing the passage of harmful microorganisms and substances into the body, while enabling the acquisition of dietary nutrients. Antibiotic growth promoters (AGP) are widely accepted as the "gold standard" of performance-enhancing feed additives, which had become integral and valuable components of modern, efficient animal production, but are now being phased out in many parts of the world. This review, therefore, examines the reported effects of AGP on the key components of gut barrier function, particularly where corresponding (positive) growth performance data were provided to indicate that any changes were beneficial, and some important trends do emerge. Certain bacterial families (e.g., Lachnospiraceae), genera (e.g., Faecalibacterium, Propionibacterium, and Ruminococcus), or species (e.g., F. prausnitzii, B. fragilis, and some Lactobacillus spp.) have been reported to increase with AGP use, are associated with improved growth performance, and show benefit across species, which may be related to their production of short-chain fatty acids (SCFA). Various studies have investigated the effects of AGP on mucus-related parameters (e.g., goblet cell size, density, and mucin mRNA expression) but these do not always seem to correlate well with the actual physical characteristics of the mucus layer(s). Surprisingly, there are little data relating to HDP or IgA, even though they have recognized benefits. There are clear AGP benefits on epithelial structure and function (e.g., nutrient digestibility), and these may (currently) provide the most reliable indicators of the efficacy of growth promoters. Data investigating effects on gut immune parameters (e.g., cell populations, cytokines, and chemokines), with corresponding growth performance, are limited and require further detailed interrogation. This review highlights both important observations related to the effects of AGP on key gut barrier components, with associated growth performance, and areas that require further investigation, thus providing an informative basis for assessing the potential of AGP alternatives.
Collapse
Affiliation(s)
- Leon J Broom
- Gut Health Consultancy, Exeter, Devon, United Kingdom.,Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, United Kingdom
| |
Collapse
|
112
|
Zhou L, Zhang M, Wang Y, Dorfman RG, Liu H, Yu T, Chen X, Tang D, Xu L, Yin Y, Pan Y, Zhou Q, Zhou Y, Yu C. Faecalibacterium prausnitzii Produces Butyrate to Maintain Th17/Treg Balance and to Ameliorate Colorectal Colitis by Inhibiting Histone Deacetylase 1. Inflamm Bowel Dis 2018; 24:1926-1940. [PMID: 29796620 DOI: 10.1093/ibd/izy182] [Citation(s) in RCA: 262] [Impact Index Per Article: 43.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Indexed: 12/11/2022]
Abstract
BACKGROUND Inflammatory bowel disease (IBD)-associated dysbiosis is characterized by a loss of Faecalibacterium prausnitzii, whose supernatant exerts an anti-inflammatory effect. However, the anti-inflammatory substances in F. prausnitzii supernatant and the mechanism in ameliorating colitis in IBD have not yet been fully investigated. METHODS Experimental colitis models were induced and evaluated by clinical examination and histopathology. Levels of cytokines and ratio of T cells were detected by enzyme-linked immunosorbent assay and flow cytometry analysis, respectively. F. prausnitzii supernatant was separated by macroporous resins. After extraction, the substances in supernatant were identified by gas chromatography-mass spectrometer. T-cell differentiation assay was conducted in vitro. Changes in signaling pathways were examined by immunoblot, immunohistochemistry, and immunofluorescent staining. RESULTS We found that the supernatant of F. prausnitzii could regulate T helper 17 cell (Th17)/regulatory T cell (Treg) differentiation. Then, we identified butyrate produced by F. prausnitzii that played the anti-inflammatory effects by inhibiting interleukin (IL)-6/signal transducer and the activator of transcription 3 (STAT3)/IL-17 pathway and promoting forkhead box protein P3 (Foxp3). Finally, we demonstrated that the target of butyrate was histone deacetylase 1 (HDAC1). CONCLUSIONS It is butyrate, instead of other substances produced by F. prausnitzii, that maintains Th17/Treg balance and exerts significant anti-inflammatory effects in colorectal colitis rodents, by inhibiting HDAC1 to promote Foxp3 and block the IL-6/STAT3/IL-17 downstream pathway. F. prausnitzii could be an option for further investigation for IBD treatment. Targeting the butyrate-HDAC1-T-cell axis offers an effective novel approach in the treatment of inflammatory disease.
Collapse
Affiliation(s)
- Lixing Zhou
- Department of Gastroenterology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Mingming Zhang
- Department of Gastroenterology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China.,Jiangsu Clinical Medical Center of Digestive Disease, Nanjing, China.,School of Life Sciences, Fudan University, Shanghai, China
| | - Yuming Wang
- Department of Gastroenterology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | | | - Hang Liu
- Department of Pharmacy, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Ting Yu
- Department of Gastroenterology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China.,Department of Gastroenterology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| | - Xiaotian Chen
- Department of Gastroenterology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Dehua Tang
- Department of Gastroenterology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Lei Xu
- Department of Gastroenterology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yuyao Yin
- Department of Gastroenterology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yida Pan
- Department of Digestive Diseases of Huashan Hospital, Shanghai, China
| | - Qian Zhou
- School of Life Sciences, Fudan University, Shanghai, China
| | - Yihua Zhou
- Key Laboratory, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Chenggong Yu
- Department of Gastroenterology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China.,Jiangsu Clinical Medical Center of Digestive Disease, Nanjing, China
| |
Collapse
|
113
|
Kalyana Chakravarthy S, Jayasudha R, Ranjith K, Dutta A, Pinna NK, Mande SS, Sharma S, Garg P, Murthy SI, Shivaji S. Alterations in the gut bacterial microbiome in fungal Keratitis patients. PLoS One 2018; 13:e0199640. [PMID: 29933394 PMCID: PMC6014669 DOI: 10.1371/journal.pone.0199640] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Accepted: 06/11/2018] [Indexed: 12/12/2022] Open
Abstract
Dysbiosis in the gut microbiome has been implicated in several diseases including auto-immune diseases, inflammatory diseases, cancers and mental disorders. Keratitis is an inflammatory disease of the eye significantly contributing to corneal blindness in the developing world. It would be worthwhile to investigate the possibility of dysbiosis in the gut microbiome being associated with Keratitis. Here, we have analyzed fungal and bacterial populations in stool samples through high-throughput sequencing of the ITS2 region for fungi and V3-V4 region of 16S rRNA gene for bacteria in healthy controls (HC, n = 31) and patients with fungal keratitis (FK, n = 32). Candida albicans (2 OTUs), Aspergillus (1 OTU) and 3 other denovo-OTUs were enriched in FK samples and an unclassified denovo-OTU was enriched in HC samples. However, the overall abundances of these ‘discriminatory’ OTUs were very low (< 0.001%) and not indicative of significant dysbiosis in the fungal community inhabiting the gut of FK patients. In contrast, the gut bacterial richness and diversity in FK patients was significantly decreased when compared to HC. 52 OTUs were significantly enriched in HC samples whereas only 5 OTUs in FK. The OTUs prominently enriched in HC were identified as Faecalibacterium prausnitzii, Bifidobacterium adolescentis, Lachnospira, Mitsuokella multacida, Bacteroides plebeius, Megasphaera and Lachnospiraceae. In FK samples, 5 OTUs affiliated to Bacteroides fragilis, Dorea, Treponema, Fusobacteriaceae, and Acidimicrobiales were significantly higher in abundance. The functional implications are that Faecalibacterium prausnitzii, an anti-inflammatory bacterium and Megasphaera, Mitsuokella multacida and Lachnospira are butyrate producers, which were enriched in HC patients, whereas Treponema and Bacteroides fragilis, which are pathogenic were abundant in FK patients, playing a potential pro-inflammatory role. Heatmap, PCoA plots and functional profiles further confirm the distinct patterns of gut bacterial composition in FK and HC samples. Our study demonstrates dysbiosis in the gut bacterial microbiomes of FK patients compared to HC. Further, based on inferred functions, it appears that dysbiosis in the gut of FK subjects is strongly associated with the disease phenotype with decrease in abundance of beneficial bacteria and increase in abundance of pro-inflammatory and pathogenic bacteria.
Collapse
Affiliation(s)
- Sama Kalyana Chakravarthy
- Jhaveri Microbiology Centre, Brien Holden Eye Research Centre, L. V. Prasad Eye Institute, Kallam Anji Reddy campus, Hyderabad, India
| | - Rajagopalaboopathi Jayasudha
- Jhaveri Microbiology Centre, Brien Holden Eye Research Centre, L. V. Prasad Eye Institute, Kallam Anji Reddy campus, Hyderabad, India
| | - Konduri Ranjith
- Jhaveri Microbiology Centre, Brien Holden Eye Research Centre, L. V. Prasad Eye Institute, Kallam Anji Reddy campus, Hyderabad, India
| | - Anirban Dutta
- Bio-Sciences R&D Division, TCS Research, Tata Consultancy Services Ltd., Pune, India
| | - Nishal Kumar Pinna
- Bio-Sciences R&D Division, TCS Research, Tata Consultancy Services Ltd., Pune, India
| | - Sharmila S. Mande
- Bio-Sciences R&D Division, TCS Research, Tata Consultancy Services Ltd., Pune, India
| | - Savitri Sharma
- Jhaveri Microbiology Centre, Brien Holden Eye Research Centre, L. V. Prasad Eye Institute, Kallam Anji Reddy campus, Hyderabad, India
| | - Prashant Garg
- Tej Kohli Cornea Institute, L. V. Prasad Eye Institute, Kallam Anji Reddy campus, Hyderabad, India
| | - Somasheila I. Murthy
- Tej Kohli Cornea Institute, L. V. Prasad Eye Institute, Kallam Anji Reddy campus, Hyderabad, India
| | - Sisinthy Shivaji
- Jhaveri Microbiology Centre, Brien Holden Eye Research Centre, L. V. Prasad Eye Institute, Kallam Anji Reddy campus, Hyderabad, India
- * E-mail:
| |
Collapse
|
114
|
The benign helminth Hymenolepis diminuta ameliorates chemically induced colitis in a rat model system. Parasitology 2018; 145:1324-1335. [DOI: 10.1017/s0031182018000896] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
AbstractThe tapeworm Hymenolepis diminuta is a model for the impact of helminth colonization on the mammalian immune system and a candidate therapeutic agent for immune mediated inflammatory diseases (IMIDs). In mice, H. diminuta protects against models of inflammatory colitis by inducing a strong type 2 immune response that is activated to expel the immature worm. Rats are the definitive host of H. diminuta, and are colonized stably and over long time periods without harming the host. Rats mount a mild type 2 immune response to H. diminuta colonization, but this response does not generally ameliorate colitis. Here we investigate the ability of different life cycle stages of H. diminuta to protect rats against a model of colitis induced through application of the haptenizing agent dinitrobenzene sulphonic acid (DNBS) directly to the colon, and monitor rat clinical health, systemic inflammation measured by TNFα and IL-1β, and the gut microbiota. We show that immature H. diminuta induces a type 2 response as measured by increased IL-4, IL-13 and IL-10 expression, but does not protect against colitis. In contrast, rats colonized with mature H. diminuta and challenged with severe colitis (two applications of DNBS) have lower inflammation and less severe clinical symptoms. This effect is not related the initial type 2 immune response. The gut microbiota is disrupted during colitis and does not appear to play an overt role in H. diminuta-mediated protection.
Collapse
|
115
|
Martín R, Chain F, Miquel S, Motta JP, Vergnolle N, Sokol H, Langella P. Using murine colitis models to analyze probiotics-host interactions. FEMS Microbiol Rev 2018; 41:S49-S70. [PMID: 28830096 DOI: 10.1093/femsre/fux035] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 06/08/2017] [Indexed: 02/07/2023] Open
Abstract
Probiotics are defined as 'live microorganisms which when administered in adequate amounts confer a health benefit on the host'. So, to consider a microorganism as a probiotic, a demonstrable beneficial effect on the health host should be shown as well as an adequate defined safety status and the capacity to survive transit through the gastrointestinal tract and to storage conditions. In this review, we present an overview of the murine colitis models currently employed to test the beneficial effect of the probiotic strains as well as an overview of the probiotics already tested. Our aim is to highlight both the importance of the adequate selection of the animal model to test the potential probiotic strains and of the value of the knowledge generated by these in vivo tests.
Collapse
Affiliation(s)
- Rebeca Martín
- INRA, Commensals and Probiotics-Host Interactions Laboratory, Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | - Florian Chain
- INRA, Commensals and Probiotics-Host Interactions Laboratory, Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | - Sylvie Miquel
- Laboratoire Microorganismes: Génome et Environnement (LMGE), UMR CNRS 6023, Université Clermont-Auvergne, 63000 Clermont-Ferrand, France
| | - Jean-Paul Motta
- Department of Biological Science, Inflammation Research Network, University of Calgary, AB T3E 4N1, Canada.,IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, F-31300 Toulouse, France
| | - Nathalie Vergnolle
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, F-31300 Toulouse, France
| | - Harry Sokol
- INRA, Commensals and Probiotics-Host Interactions Laboratory, Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France.,Sorbonne University - Université Pierre et Marie Curie (UPMC), 75252 Paris, France.,Institut National de la Santé et de la Recherche Médicale (INSERM) Equipe de Recherche Labélisée (ERL) 1157, Avenir Team Gut Microbiota and Immunity, 75012 Paris, France.,Department of Gastroenterology, Saint Antoine Hospital, Assistance Publique - Hopitaux de Paris, UPMC, 75012 Paris, France
| | - Philippe Langella
- INRA, Commensals and Probiotics-Host Interactions Laboratory, Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| |
Collapse
|
116
|
Yu YJ, Wang XH, Fan GC. Versatile effects of bacterium-released membrane vesicles on mammalian cells and infectious/inflammatory diseases. Acta Pharmacol Sin 2018; 39:514-533. [PMID: 28858295 PMCID: PMC5888691 DOI: 10.1038/aps.2017.82] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 05/10/2017] [Indexed: 01/18/2023] Open
Abstract
Gram-negative bacterium-released outer-membrane vesicles (OMVs) and Gram-positive bacterium-released membrane vesicles (MVs) share significant similarities with mammalian cell-derived MVs (eg, microvesicles and exosomes) in terms of structure and their biological activities. Recent studies have revealed that bacterial OMVs/MVs could (1) interact with immune cells to regulate inflammatory responses, (2) transport virulence factors (eg, enzymes, DNA and small RNAs) to host cells and result in cell injury, (3) enhance barrier function by stimulating the expression of tight junction proteins in intestinal epithelial cells, (4) upregulate the expression of endothelial cell adhesion molecules, and (5) serve as natural nanocarriers for immunogenic antigens, enzyme support and drug delivery. In addition, OMVs/MVs can enter the systemic circulation and induce a variety of immunological and metabolic responses. This review highlights the recent advances in the understanding of OMV/MV biogenesis and their compositional remodeling. In addition, interactions between OMVs/MVs and various types of mammalian cells (ie, immune cells, epithelial cells, and endothelial cells) and their pathological/preventive effects on infectious/inflammatory diseases are summarized. Finally, methods for engineering OMVs/MVs and their therapeutic potential are discussed.
Collapse
Affiliation(s)
- You-jiang Yu
- Medical College of Yangzhou Polytechnic College, Yangzhou 225009, China
| | - Xiao-hong Wang
- Department of Pharmacology and Cell Biophysics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Guo-Chang Fan
- Department of Pharmacology and Cell Biophysics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| |
Collapse
|
117
|
Barone M, Chain F, Sokol H, Brigidi P, Bermúdez-Humarán LG, Langella P, Martín R. A Versatile New Model of Chemically Induced Chronic Colitis Using an Outbred Murine Strain. Front Microbiol 2018; 9:565. [PMID: 29636738 PMCID: PMC5881104 DOI: 10.3389/fmicb.2018.00565] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 03/12/2018] [Indexed: 01/01/2023] Open
Abstract
Murine colitis models are crucial tools for understanding intestinal homeostasis and inflammation. However, most current models utilize a highly inbred strain of mice, and often only one sex is employed to limit bias. This targeted approach, which in itself is biased, means that murine genetic diversity and sex-related differences are ignored, making it even more difficult to extend findings to humans, who are highly heterogeneous. Furthermore, most models do not examine the chronic form of colitis, an important fact taking into account the chronic nature of the inflammatory bowel diseases (IBD). Here, we attempted to create a more realistic murine colitis model by addressing these three issues. Using chemically induced chronic colon inflammation in an outbred strain of mice (RjOrl:SWISS [CD-1]), we (i) mimicked the relapsing nature of the disease, (ii) better represented normal genetic variability, and (iii) employed both female and male mice. Colitis was induced by intrarectal administration of dinitrobenzene sulfonic acid (DNBS). After a recovery period and 3 days before the mice were euthanized, colitis was reactivated by a second administration of DNBS. Protocol length was 24 days. Colitis severity was assessed using body mass, macroscopic scores, and histological scores. Myeloperoxidase (MPO) activity, cytokine levels, and lymphocyte populations were also characterized. Our results show that the intrarectal administration of DNBS effectively causes colitis in both female and male CD-1 mice in a dose-dependent manner, as reflected by loss of body mass, macroscopic scores and histological scores. Furthermore, colon cytokine levels and mesenteric lymph node characteristics indicate that this model involves immune system activation. Although some variables were sex-specific, most of the results support including both females and males in the model. Our ultimate goal is to make this model available to researchers for testing candidate anti-inflammatory agents, such as classical or next-generation probiotics; we also aim for the results to be more easily transferrable to human trials.
Collapse
Affiliation(s)
- Monica Barone
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Florian Chain
- Commensals and Probiotics-Host Interactions Laboratory, Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Harry Sokol
- Commensals and Probiotics-Host Interactions Laboratory, Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France.,Sorbonne University - Université Pierre et Marie Curie, Paris, France.,Avenir Team Gut Microbiota and Immunity, Institut National de la Santé et de la Recherche Médicale, Equipe de Recherche Labélisée 1157, Paris, France.,Department of Gastroenterology, Saint Antoine Hospital, Assistance Publique-Hôpitaux de Paris, UPMC, Paris, France
| | - Patrizia Brigidi
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Luis G Bermúdez-Humarán
- Commensals and Probiotics-Host Interactions Laboratory, Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Philippe Langella
- Commensals and Probiotics-Host Interactions Laboratory, Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Rebeca Martín
- Commensals and Probiotics-Host Interactions Laboratory, Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| |
Collapse
|
118
|
Martín R, Bermúdez-Humarán LG, Langella P. Searching for the Bacterial Effector: The Example of the Multi-Skilled Commensal Bacterium Faecalibacterium prausnitzii. Front Microbiol 2018; 9:346. [PMID: 29559959 PMCID: PMC5845625 DOI: 10.3389/fmicb.2018.00346] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 02/13/2018] [Indexed: 01/02/2023] Open
Abstract
Faecalibacterium prausnitzii represents approximately 5% of the total fecal microbiota in healthy adults being one of the most abundant bacterium in the human intestinal microbiota of healthy adults. Furthermore, this bacterium has been proposed to be a sensor and a major actor of the human intestinal health because of its importance in the gut ecosystem. In this context, F. prausnitzii population levels have been found to be reduced in patients suffering from several syndromes and diseases such as inflammatory bowel diseases. These diseases are characterized by a breakage of the intestinal homeostasis called dysbiosis and the use of F. prausnitzii as a next generation probiotic (also called live biotherapeutics) has been proposed as a natural tool to restore such dysbiosis within the gut. Nevertheless, despite the potential importance of this bacterium in human health, little is known about its main effectors underlying its beneficial effects. In this perspective note, we aim to present the actual state in the research about F. prausnitzii effectors and the future milestones in this field.
Collapse
Affiliation(s)
- Rebeca Martín
- National Institute of Agricultural Research, Commensals and Probiotics-Host Interactions Laboratory, Micalis Institute, AgroParisTech, Paris-Sud University, Jouy-en-Josas, France
| | - Luis G Bermúdez-Humarán
- National Institute of Agricultural Research, Commensals and Probiotics-Host Interactions Laboratory, Micalis Institute, AgroParisTech, Paris-Sud University, Jouy-en-Josas, France
| | - Philippe Langella
- National Institute of Agricultural Research, Commensals and Probiotics-Host Interactions Laboratory, Micalis Institute, AgroParisTech, Paris-Sud University, Jouy-en-Josas, France
| |
Collapse
|
119
|
Laserna-Mendieta EJ, Clooney AG, Carretero-Gomez JF, Moran C, Sheehan D, Nolan JA, Hill C, Gahan CGM, Joyce SA, Shanahan F, Claesson MJ. Determinants of Reduced Genetic Capacity for Butyrate Synthesis by the Gut Microbiome in Crohn's Disease and Ulcerative Colitis. J Crohns Colitis 2018; 12:204-216. [PMID: 29373727 DOI: 10.1093/ecco-jcc/jjx137] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 10/05/2017] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIMS Alterations in short chain fatty acid metabolism, particularly butyrate, have been reported in inflammatory bowel disease, but results have been conflicting because of small study numbers and failure to distinguish disease type, activity or other variables such as diet. We performed a comparative assessment of the capacity of the microbiota for butyrate synthesis, by quantifying butyryl-CoA:acetate CoA-transferase [BCoAT] gene content in stool from patients with Crohn's disease [CD; n = 71], ulcerative colitis [UC; n = 58] and controls [n = 75], and determined whether it was related to active vs inactive inflammation, microbial diversity, and composition and/or dietary habits. METHODS BCoAT gene content was quantified by quantitative polymerase chain reaction [qPCR]. Disease activity was assessed clinically and faecal calprotectin concentration measured. Microbial composition was determined by sequencing 16S rRNA gene. Dietary data were collected using an established food frequency questionnaire. RESULTS Reduced butyrate-synthetic capacity was found in patients with active and inactive CD [p < 0.001 and p < 0.01, respectively], but only in active UC [p < 0.05]. In CD, low BCoAT gene content was associated with ileal location, stenotic behaviour, increased inflammation, lower microbial diversity, greater microbiota compositional change, and decreased butyrogenic taxa. Reduced BCoAT gene content in patients with CD was linked with a different regimen characterised by lower dietary fibre. CONCLUSIONS Reduced butyrate-synthetic capacity of the microbiota is more evident in CD than UC and may relate to reduced fibre intake. The results suggest that simple replacement of butyrate per se may be therapeutically inadequate, whereas manipulation of microbial synthesis, perhaps by dietary means, may be more appropriate.
Collapse
Affiliation(s)
- Emilio J Laserna-Mendieta
- APC Microbiome Institute, University College Cork, Cork, Ireland.,School of Microbiology, University College Cork, Cork, Ireland
| | - Adam G Clooney
- APC Microbiome Institute, University College Cork, Cork, Ireland.,School of Microbiology, University College Cork, Cork, Ireland
| | | | - Carthage Moran
- APC Microbiome Institute, University College Cork, Cork, Ireland.,School of Medicine, University College Cork, Cork, Ireland
| | - Donal Sheehan
- APC Microbiome Institute, University College Cork, Cork, Ireland.,School of Medicine, University College Cork, Cork, Ireland
| | - James A Nolan
- APC Microbiome Institute, University College Cork, Cork, Ireland.,School of Microbiology, University College Cork, Cork, Ireland
| | - Colin Hill
- APC Microbiome Institute, University College Cork, Cork, Ireland.,School of Microbiology, University College Cork, Cork, Ireland
| | - Cormac G M Gahan
- APC Microbiome Institute, University College Cork, Cork, Ireland.,School of Microbiology, University College Cork, Cork, Ireland.,School of Pharmacy, University College Cork, Cork, Ireland
| | - Susan A Joyce
- APC Microbiome Institute, University College Cork, Cork, Ireland.,School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
| | - Fergus Shanahan
- APC Microbiome Institute, University College Cork, Cork, Ireland.,School of Medicine, University College Cork, Cork, Ireland
| | - Marcus J Claesson
- APC Microbiome Institute, University College Cork, Cork, Ireland.,School of Microbiology, University College Cork, Cork, Ireland
| |
Collapse
|
120
|
Jiang Z, Zhao M, Zhang H, Li Y, Liu M, Feng F. Antimicrobial Emulsifier-Glycerol Monolaurate Induces Metabolic Syndrome, Gut Microbiota Dysbiosis, and Systemic Low-Grade Inflammation in Low-Fat Diet Fed Mice. Mol Nutr Food Res 2018; 62. [PMID: 29131494 DOI: 10.1002/mnfr.201700547] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2017] [Revised: 09/27/2017] [Indexed: 12/12/2022]
Abstract
SCOPE Glycerol monolaurate (GML) is widely consumed worldwide in the food industry and is considered safe, but for chronic diseases, supporting scientific data remain sparse. This study investigates whether dietary GML induces metabolic syndrome, gut microbiota dysbiosis, and systemic low-grade inflammation. METHODS AND RESULTS GML-induced occurrence of metabolic syndrome, gut microbiota alterations, and systemic low-grade inflammation are investigated. The results demonstrate that GML induced metabolic syndrome by significantly increasing the body weight, weight gain, food intake, body fat, fat droplet size and percentage of epididymal fat, serum triglycerides (TG), LDL, and atherogenic index, and decreasing the body muscle ratio, liver weight, and HDL, compared to the control (CON) group. Meanwhile, GML significantly changed the β-diversity and composition of gut microbiota and upregulated the circulating levels of serum LPS, IL-1β, IL-6, and TNF-α. Importantly, GML significantly decreased Akkermansia muciniphila and Lupinus luteus, and increased Bacteroides acidifaciens, Escherichia coli and the microbial DNA abundance of the ten predicated metabolism pathways involved in carbohydrate, amino acid, and lipid metabolism. CONCLUSION Our results indicate that relatively low-dose GML consumption promotes metabolic syndrome, gut microbiota dysbiosis, and systemic low-grade inflammation, thereby calling for a reassessment of GML usage.
Collapse
Affiliation(s)
- Zengliang Jiang
- College of Biosystems Engineering and Food Science, Fuli Institute for Food Science, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang Engineering Center for Food Technology and Equipment, Zhejiang University, Hangzhou, China
| | - Minjie Zhao
- College of Biosystems Engineering and Food Science, Fuli Institute for Food Science, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang Engineering Center for Food Technology and Equipment, Zhejiang University, Hangzhou, China
| | - Hui Zhang
- College of Biosystems Engineering and Food Science, Fuli Institute for Food Science, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang Engineering Center for Food Technology and Equipment, Zhejiang University, Hangzhou, China
| | - Yang Li
- College of Biosystems Engineering and Food Science, Fuli Institute for Food Science, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang Engineering Center for Food Technology and Equipment, Zhejiang University, Hangzhou, China
| | - Mengyun Liu
- College of Biosystems Engineering and Food Science, Fuli Institute for Food Science, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang Engineering Center for Food Technology and Equipment, Zhejiang University, Hangzhou, China
| | - Fengqin Feng
- College of Biosystems Engineering and Food Science, Fuli Institute for Food Science, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang Engineering Center for Food Technology and Equipment, Zhejiang University, Hangzhou, China
| |
Collapse
|
121
|
Allouche R, Dupont S, Charriau A, Gervais P, Beney L, Chambin O. Optimized tableting for extremely oxygen-sensitive probiotics using direct compression. Int J Pharm 2018; 538:14-20. [PMID: 29307771 DOI: 10.1016/j.ijpharm.2018.01.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 12/19/2017] [Accepted: 01/03/2018] [Indexed: 01/25/2023]
Abstract
Faecalibacterium prausnitzii was previously recognized for its intestinal anti-inflammatory activities and it has been shown less abundant in patients with chronic intestinal diseases. However, the main problems encountered in the use of this interesting anaerobic microorganism are firstly its high sensitivity to the oxygen and secondly, its ability to reach the large intestine alive as targeted site. The aim of this study was to investigate the effect of direct compression on the viability of this probiotic strain after different compression pressure and storage using three different excipients (MCC, HPMC and HPMCP). The effect of compression process on cell viability was studied and a strategy was proposed to improve probiotic viability. Results showed that cell viability decreased almost linearly with compression pressure. MCC and HPMC seemed the most favorable carriers and after storage, each tablet exhibited a survival above108 CFU. Storage stability was obtained with a pressure of 201 MPa after 28 days at 25 °C, in anaerobic condition and with 11% relative humidity. Compression after a pre-consolidated stage improved clearly the survival rate due to lower temperature increase and lower shearing force. Thus, direct compression seems to be suitable in producing probiotics tablets with extremely oxygen-sensitive strains, and could provide sufficient protection during storage to expect therapeutic efficiency.
Collapse
Affiliation(s)
- Rania Allouche
- Université de Bourgogne Franche-Comté, AgroSup Dijon, PAM UMR A 02.102, F-21000, Dijon, France
| | - Sébastien Dupont
- Université de Bourgogne Franche-Comté, AgroSup Dijon, PAM UMR A 02.102, F-21000, Dijon, France
| | - Alexandre Charriau
- Université de Bourgogne Franche-Comté, AgroSup Dijon, PAM UMR A 02.102, F-21000, Dijon, France
| | - Patrick Gervais
- Université de Bourgogne Franche-Comté, AgroSup Dijon, PAM UMR A 02.102, F-21000, Dijon, France
| | - Laurent Beney
- Université de Bourgogne Franche-Comté, AgroSup Dijon, PAM UMR A 02.102, F-21000, Dijon, France
| | - Odile Chambin
- Université de Bourgogne Franche-Comté, AgroSup Dijon, PAM UMR A 02.102, F-21000, Dijon, France; Department of Pharmaceutical Technology, Health Sciences Faculty, Université de Bourgogne Franche-Comté, 7 Bd Jeanne d'Arc, F-21079, Dijon, France.
| |
Collapse
|
122
|
Zhang SL, Wang SN, Miao CY. Influence of Microbiota on Intestinal Immune System in Ulcerative Colitis and Its Intervention. Front Immunol 2017; 8:1674. [PMID: 29234327 PMCID: PMC5712343 DOI: 10.3389/fimmu.2017.01674] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Accepted: 11/14/2017] [Indexed: 01/07/2023] Open
Abstract
Ulcerative colitis (UC) is an inflammatory bowel disease (IBD) with chronic and recurrent characteristics caused by multiple reasons. Although the pathogenic factors have not been clarified yet, recent studies have demonstrated that intestinal microbiota plays a major role in UC, especially in the immune system. This review focuses on the description of several major microbiota communities that affect UC and their interactions with the host. In this review, eight kinds of microbiota that are highly related to IBD, including Faecalibacterium prausnitzii, Clostridium clusters IV and XIVa, Bacteroides, Roseburia species, Eubacterium rectale, Escherichia coli, Fusobacterium, and Candida albicans are demonstrated on the changes in amount and roles in the onset and progression of IBD. In addition, potential therapeutic targets for UC involved in the regulation of microbiota, including NLRPs, vitamin D receptor as well as secreted proteins, are discussed in this review.
Collapse
Affiliation(s)
- Sai-Long Zhang
- Department of Pharmacology, Second Military Medical University, Shanghai, China
| | - Shu-Na Wang
- Department of Pharmacology, Second Military Medical University, Shanghai, China
| | - Chao-Yu Miao
- Department of Pharmacology, Second Military Medical University, Shanghai, China
| |
Collapse
|
123
|
Chung WSF, Meijerink M, Zeuner B, Holck J, Louis P, Meyer AS, Wells JM, Flint HJ, Duncan SH. Prebiotic potential of pectin and pectic oligosaccharides to promote anti-inflammatory commensal bacteria in the human colon. FEMS Microbiol Ecol 2017; 93:4331632. [DOI: 10.1093/femsec/fix127] [Citation(s) in RCA: 136] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 09/29/2017] [Indexed: 01/16/2023] Open
|
124
|
El Hage R, Hernandez-Sanabria E, Van de Wiele T. Emerging Trends in "Smart Probiotics": Functional Consideration for the Development of Novel Health and Industrial Applications. Front Microbiol 2017; 8:1889. [PMID: 29033923 PMCID: PMC5626839 DOI: 10.3389/fmicb.2017.01889] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 09/14/2017] [Indexed: 12/12/2022] Open
Abstract
The link between gut microbiota and human health is well-recognized and described. This ultimate impact on the host has contributed to explain the mutual dependence between humans and their gut bacteria. Gut microbiota can be manipulated through passive or active strategies. The former includes diet, lifestyle, and environment, while the latter comprise antibiotics, pre- and probiotics. Historically, conventional probiotic strategies included a phylogenetically limited diversity of bacteria and some yeast strains. However, biotherapeutic strategies evolved in the last years with the advent of fecal microbiota transplant (FMT), successfully applied for treating CDI, IBD, and other diseases. Despite the positive outcomes, long-term effects resulting from the uncharacterized nature of FMT are not sufficiently studied. Thus, developing strategies to simulate the FMT, using characterized gut colonizers with identified phylogenetic diversity, may be a promising alternative. As the definition of probiotics states that the microorganism should have beneficial effects on the host, several bacterial species with proven efficacy have been considered next generation probiotics. Non-conventional candidate strains include Akkermansia muciniphila, Faecalibacterium prausnitzii, Bacteroides fragilis, and members of the Clostridia clusters IV, XIVa, and XVIII. However, viable intestinal delivery is one of the current challenges, due to their stringent survival conditions. In this review, we will cover current perspectives on the development and assessment of next generation probiotics and the approaches that industry and stakeholders must consider for a successful outcome.
Collapse
Affiliation(s)
| | | | - Tom Van de Wiele
- Center for Microbial Ecology and Technology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| |
Collapse
|
125
|
Eichele DD, Kharbanda KK. Dextran sodium sulfate colitis murine model: An indispensable tool for advancing our understanding of inflammatory bowel diseases pathogenesis. World J Gastroenterol 2017; 23:6016-6029. [PMID: 28970718 PMCID: PMC5597494 DOI: 10.3748/wjg.v23.i33.6016] [Citation(s) in RCA: 514] [Impact Index Per Article: 73.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 07/07/2017] [Accepted: 08/02/2017] [Indexed: 02/06/2023] Open
Abstract
Inflammatory bowel diseases (IBD), including Crohn’s disease and ulcerative colitis, are complex diseases that result from the chronic dysregulated immune response in the gastrointestinal tract. The exact etiology is not fully understood, but it is accepted that it occurs when an inappropriate aggressive inflammatory response in a genetically susceptible host due to inciting environmental factors occurs. To investigate the pathogenesis and etiology of human IBD, various animal models of IBD have been developed that provided indispensable insights into the histopathological and morphological changes as well as factors associated with the pathogenesis of IBD and evaluation of therapeutic options in the last few decades. The most widely used experimental model employs dextran sodium sulfate (DSS) to induce epithelial damage. The DSS colitis model in IBD research has advantages over other various chemically induced experimental models due to its rapidity, simplicity, reproducibility and controllability. In this manuscript, we review the newer publicized advances of research in murine colitis models that focus upon the disruption of the barrier function of the intestine, effects of mucin on the development of colitis, alterations found in microbial balance and resultant changes in the metabolome specifically in the DSS colitis murine model and its relation to the pathogenesis of IBD.
Collapse
Affiliation(s)
- Derrick D Eichele
- Department of Internal Medicine, Nebraska Medical Center, Omaha, NE 68198, United States
| | - Kusum K Kharbanda
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE 68105, United States
- Department of Internal Medicine, Nebraska Medical Center, Omaha, NE 68198, United States
- Department of Biochemistry and Molecular Biology, Nebraska Medical Center, Omaha, NE 68198, United States
| |
Collapse
|
126
|
Salaga M, Mokrowiecka A, Jacenik D, Cygankiewicz AI, Malecka-Panas E, Kordek R, Krajewska WM, Sobocinska MK, Kamysz E, Fichna J. Systemic Administration of Sialorphin Attenuates Experimental Colitis in Mice via Interaction With Mu and Kappa Opioid Receptors. J Crohns Colitis 2017; 11:988-998. [PMID: 28333341 DOI: 10.1093/ecco-jcc/jjx043] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 03/17/2017] [Indexed: 02/08/2023]
Abstract
BACKGROUND AND AIMS Pharmacological treatment and/or maintenance of remission in inflammatory bowel disease [IBD] is currently one of the biggest challenges in the field of gastroenterology. Here we aimed to assess the anti-inflammatory effect and the mechanism of action of sialorphin, the natural blocker of the endogenous opioid peptide-degrading enzymes neprilysin [NEP] and aminopeptidase N [APN], in mouse models of IBD and the changes in the expression of these enzymes in IBD patients. METHODS We used two models of experimental colitis in mice [2,4,6-trinitrobenzene sulphonic acid [TNBS]- and dextran sulphate sodium [DSS]-induced]. Macroscopic score, ulcer score, colonic wall thickness, and myeloperoxidase [MPO] activity were recorded. Additionally, we measured the expression of NEP and APN in the colon of IBD patients and healthy controls. RESULTS We showed that sialorphin attenuated acute, semichronic, and relapsing TNBS-induced colitis in mice after systemic administration, and its anti-inflammatory action is associated with mu and kappa opioid receptors. CONCLUSIONS We show that indirect stimulation of opioid receptors by the blockade of NEP and APN is a promising pharmacological strategy for the treatment of IBD, and may become of greater importance than the use of classical opioid agonists.
Collapse
Affiliation(s)
- M Salaga
- Department Biochemistry, Medical University of Lodz, Lodz, Poland
| | - A Mokrowiecka
- Department of Digestive Tract Diseases, Medical University of Lodz, Lodz, Poland
| | - D Jacenik
- Department of Cytobiochemistry, University of Lodz, Lodz, Poland
| | - A I Cygankiewicz
- Department of Molecular Biotechnology, University of Gdansk, Gdansk, Poland
| | - E Malecka-Panas
- Department of Digestive Tract Diseases, Medical University of Lodz, Lodz, Poland
| | - R Kordek
- Department of Pathology, Faculty of Medicine,Lodz, Poland
| | - W M Krajewska
- Department of Cytobiochemistry, University of Lodz, Lodz, Poland
| | - M K Sobocinska
- Department of Molecular Biotechnology, University of Gdansk, Gdansk, Poland
| | - E Kamysz
- Department of Molecular Biotechnology, University of Gdansk, Gdansk, Poland
| | - J Fichna
- Department Biochemistry, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
127
|
Salaga M, Mokrowiecka A, Zielinska M, Malecka-Panas E, Kordek R, Kamysz E, Fichna J. New Peptide Inhibitor of Dipeptidyl Peptidase IV, EMDB-1 Extends the Half-Life of GLP-2 and Attenuates Colitis in Mice after Topical Administration. J Pharmacol Exp Ther 2017; 363:92-103. [PMID: 28724693 DOI: 10.1124/jpet.117.242586] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 07/12/2017] [Indexed: 12/20/2022] Open
Abstract
Protease inhibition has become a possible new approach in inflammatory bowel disease (IBD) therapy. A serine exopeptidase, dipeptidyl peptidase IV (DPP IV), is responsible for the inactivation of incretin hormone, glucagon-like peptide 2 (GLP-2), a potent stimulator of intestinal epithelium regeneration and growth. Recently, we showed that the novel peptide analog of endomorphin-2, Tyr-Pro-D-ClPhe-Phe-NH2 (EMDB-1) is a potent blocker of DPP IV and has an inhibitory effect on gastrointestinal (GI) smooth muscle contractility. The aim of this study was to characterize the anti-inflammatory effect and mechanism of action of EMDB-1 in the mouse GI tract. We used two models of experimental colitis (induced by TNBS and DSS). The anti-inflammatory effect of EMDB-1 was assessed by the determination of macroscopic score, ulcer score, colonic wall thickness, as well as myeloperoxidase activity. Additionally, we measured the expression of GLP-2, GLP2R, and DPP IV in the colon of control and colitic animals treated with the test compound. The expression of GLP-2 and GLP2R in the serum and colon of IBD patients and healthy control subjects has been assessed. We showed that EMDB-1 elevates the half-life of GLP-2 in vitro and attenuates acute, semichronic, and relapsing TNBS as well as DSS-induced colitis in mice after topical administration. The anti-inflammatory action of EMDB-1 is associated with changes in the level of colonic GLP-2 but not DPP IV expression. Our results validate DPP IV as a pharmacological target for the anti-IBD drugs, and its inhibitors based on natural substrates, such as EMDB-1, have the potential to become valuable anti-inflammatory therapeutic agents.
Collapse
Affiliation(s)
- Maciej Salaga
- Department of Biochemistry, Department of Digestive Tract Diseases, Department of Pathology, Faculty of Medicine, Medical University of Lodz, Lodz, Poland (M.S., A.M., M.Z., E.M.P., R.K., J.F.); and Department of Molecular Biotechnology, Faculty of Chemistry, University of Gdansk, Gdansk, Poland (E.K.)
| | - Anna Mokrowiecka
- Department of Biochemistry, Department of Digestive Tract Diseases, Department of Pathology, Faculty of Medicine, Medical University of Lodz, Lodz, Poland (M.S., A.M., M.Z., E.M.P., R.K., J.F.); and Department of Molecular Biotechnology, Faculty of Chemistry, University of Gdansk, Gdansk, Poland (E.K.)
| | - Marta Zielinska
- Department of Biochemistry, Department of Digestive Tract Diseases, Department of Pathology, Faculty of Medicine, Medical University of Lodz, Lodz, Poland (M.S., A.M., M.Z., E.M.P., R.K., J.F.); and Department of Molecular Biotechnology, Faculty of Chemistry, University of Gdansk, Gdansk, Poland (E.K.)
| | - Ewa Malecka-Panas
- Department of Biochemistry, Department of Digestive Tract Diseases, Department of Pathology, Faculty of Medicine, Medical University of Lodz, Lodz, Poland (M.S., A.M., M.Z., E.M.P., R.K., J.F.); and Department of Molecular Biotechnology, Faculty of Chemistry, University of Gdansk, Gdansk, Poland (E.K.)
| | - Radzislaw Kordek
- Department of Biochemistry, Department of Digestive Tract Diseases, Department of Pathology, Faculty of Medicine, Medical University of Lodz, Lodz, Poland (M.S., A.M., M.Z., E.M.P., R.K., J.F.); and Department of Molecular Biotechnology, Faculty of Chemistry, University of Gdansk, Gdansk, Poland (E.K.)
| | - Elzbieta Kamysz
- Department of Biochemistry, Department of Digestive Tract Diseases, Department of Pathology, Faculty of Medicine, Medical University of Lodz, Lodz, Poland (M.S., A.M., M.Z., E.M.P., R.K., J.F.); and Department of Molecular Biotechnology, Faculty of Chemistry, University of Gdansk, Gdansk, Poland (E.K.)
| | - Jakub Fichna
- Department of Biochemistry, Department of Digestive Tract Diseases, Department of Pathology, Faculty of Medicine, Medical University of Lodz, Lodz, Poland (M.S., A.M., M.Z., E.M.P., R.K., J.F.); and Department of Molecular Biotechnology, Faculty of Chemistry, University of Gdansk, Gdansk, Poland (E.K.)
| |
Collapse
|
128
|
Martín R, Miquel S, Benevides L, Bridonneau C, Robert V, Hudault S, Chain F, Berteau O, Azevedo V, Chatel JM, Sokol H, Bermúdez-Humarán LG, Thomas M, Langella P. Functional Characterization of Novel Faecalibacterium prausnitzii Strains Isolated from Healthy Volunteers: A Step Forward in the Use of F. prausnitzii as a Next-Generation Probiotic. Front Microbiol 2017; 8:1226. [PMID: 28713353 PMCID: PMC5492426 DOI: 10.3389/fmicb.2017.01226] [Citation(s) in RCA: 249] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 06/16/2017] [Indexed: 12/16/2022] Open
Abstract
Faecalibacterium prausnitzii is a major member of the Firmicutes phylum and one of the most abundant bacteria in the healthy human microbiota. F. prausnitzii depletion has been reported in several intestinal disorders, and more consistently in Crohn's disease (CD) patients. Despite its importance in human health, only few microbiological studies have been performed to isolate novel F. prausnitzii strains in order to better understand the biodiversity and physiological diversity of this beneficial commensal species. In this study, we described a protocol to isolate novel F. prausnitzii strains from feces of healthy volunteers as well as a deep molecular and metabolic characterization of these isolated strains. These F. prausnitzii strains were classified in two phylogroups and three clusters according to 16S rRNA sequences and results support that they would belong to two different genomospecies or genomovars as no genome sequencing has been performed in this work. Differences in enzymes production, antibiotic resistance and immunomodulatory properties were found to be strain-dependent. So far, all F. prausnitzii isolates share some characteristic such as (i) the lack of epithelial cells adhesion, plasmids, anti-microbial, and hemolytic activity and (ii) the presence of DNAse activity. Furthermore, Short Chain Fatty Acids (SCFA) production was assessed for the novel isolates as these products influence intestinal homeostasis. Indeed, the butyrate production has been correlated to the capacity to induce IL-10, an anti-inflammatory cytokine, in peripheral blood mononuclear cells (PBMC) but not to the ability to block IL-8 secretion in TNF-α-stimulated HT-29 cells, reinforcing the hypothesis of a complex anti-inflammatory pathway driven by F. prausnitzii. Altogether, our results suggest that some F. prausnitzii strains could represent good candidates as next-generation probiotic.
Collapse
Affiliation(s)
- Rebeca Martín
- Commensals and Probiotics-Host Interactions Laboratory, Micalis Institute, Institut National de la Recherche Agronomique, AgroParisTech, Université Paris-SaclayJouy-en-Josas, France
| | - Sylvie Miquel
- Commensals and Probiotics-Host Interactions Laboratory, Micalis Institute, Institut National de la Recherche Agronomique, AgroParisTech, Université Paris-SaclayJouy-en-Josas, France.,Université Clermont Auvergne, Centre National de la Recherche Scientifique UMR 6023 Laboratoire Microorganismes: Génome et EnvironnementClermont-Ferrand, France
| | - Leandro Benevides
- Commensals and Probiotics-Host Interactions Laboratory, Micalis Institute, Institut National de la Recherche Agronomique, AgroParisTech, Université Paris-SaclayJouy-en-Josas, France.,Department of General Biology, Federal University of Minas GeraisBelo Horizonte, Brazil
| | - Chantal Bridonneau
- Commensals and Probiotics-Host Interactions Laboratory, Micalis Institute, Institut National de la Recherche Agronomique, AgroParisTech, Université Paris-SaclayJouy-en-Josas, France
| | - Véronique Robert
- Commensals and Probiotics-Host Interactions Laboratory, Micalis Institute, Institut National de la Recherche Agronomique, AgroParisTech, Université Paris-SaclayJouy-en-Josas, France
| | - Sylvie Hudault
- Commensals and Probiotics-Host Interactions Laboratory, Micalis Institute, Institut National de la Recherche Agronomique, AgroParisTech, Université Paris-SaclayJouy-en-Josas, France
| | - Florian Chain
- Commensals and Probiotics-Host Interactions Laboratory, Micalis Institute, Institut National de la Recherche Agronomique, AgroParisTech, Université Paris-SaclayJouy-en-Josas, France
| | - Olivier Berteau
- Commensals and Probiotics-Host Interactions Laboratory, Micalis Institute, Institut National de la Recherche Agronomique, AgroParisTech, Université Paris-SaclayJouy-en-Josas, France
| | - Vasco Azevedo
- Department of General Biology, Federal University of Minas GeraisBelo Horizonte, Brazil
| | - Jean M Chatel
- Commensals and Probiotics-Host Interactions Laboratory, Micalis Institute, Institut National de la Recherche Agronomique, AgroParisTech, Université Paris-SaclayJouy-en-Josas, France
| | - Harry Sokol
- Commensals and Probiotics-Host Interactions Laboratory, Micalis Institute, Institut National de la Recherche Agronomique, AgroParisTech, Université Paris-SaclayJouy-en-Josas, France.,AVENIR Team Gut Microbiota and Immunity Equipe de Recherche Labélisée (ERL), Institut National de la Santé et de la Recherche Médicale U1157/UMR7203, Faculté de Médecine Saint-Antoine, Université Pierre et Marie CurieParis, France.,Service de Gastroentérologie, Hôpital Saint-Antoine, Assistance Publique-Hôpitaux de ParisParis, France
| | - Luis G Bermúdez-Humarán
- Commensals and Probiotics-Host Interactions Laboratory, Micalis Institute, Institut National de la Recherche Agronomique, AgroParisTech, Université Paris-SaclayJouy-en-Josas, France
| | - Muriel Thomas
- Commensals and Probiotics-Host Interactions Laboratory, Micalis Institute, Institut National de la Recherche Agronomique, AgroParisTech, Université Paris-SaclayJouy-en-Josas, France
| | - Philippe Langella
- Commensals and Probiotics-Host Interactions Laboratory, Micalis Institute, Institut National de la Recherche Agronomique, AgroParisTech, Université Paris-SaclayJouy-en-Josas, France
| |
Collapse
|
129
|
Lopez-Siles M, Duncan SH, Garcia-Gil LJ, Martinez-Medina M. Faecalibacterium prausnitzii: from microbiology to diagnostics and prognostics. THE ISME JOURNAL 2017; 11:841-852. [PMID: 28045459 PMCID: PMC5364359 DOI: 10.1038/ismej.2016.176] [Citation(s) in RCA: 460] [Impact Index Per Article: 65.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 10/15/2016] [Accepted: 11/10/2016] [Indexed: 12/13/2022]
Abstract
There is an increasing interest in Faecalibacterium prausnitzii, one of the most abundant bacterial species found in the gut, given its potentially important role in promoting gut health. Although some studies have phenotypically characterized strains of this species, it remains a challenge to determine which factors have a key role in maintaining the abundance of this bacterium in the gut. Besides, phylogenetic analysis has shown that at least two different F. prausnitzii phylogroups can be found within this species and their distribution is different between healthy subjects and patients with gut disorders. It also remains unknown whether or not there are other phylogroups within this species, and also if other Faecalibacterium species exist. Finally, many studies have shown that F. prausnitzii abundance is reduced in different intestinal disorders. It has been proposed that F. prausnitzii monitoring may therefore serve as biomarker to assist in gut diseases diagnostics. In this mini-review, we aim to serve as an overview of F. prausnitzii phylogeny, ecophysiology and diversity. In addition, strategies to modulate the abundance of F. prausnitzii in the gut as well as its application as a biomarker for diagnostics and prognostics of gut diseases are discussed. This species may be a useful potential biomarker to assist in ulcerative colitis and Crohn's disease discrimination.
Collapse
Affiliation(s)
- Mireia Lopez-Siles
- Laboratori de Microbiologia Molecular, Departament de Biologia, Universitat de Girona, Girona, Spain
| | - Sylvia H Duncan
- Microbiology Group, Rowett Institute of Nutrition and Health, University of Aberdeen, Aberdeen, UK
| | - L Jesús Garcia-Gil
- Laboratori de Microbiologia Molecular, Departament de Biologia, Universitat de Girona, Girona, Spain
- Laboratori de Microbiologia Molecular, Departament de Biologia, Universitat de Girona, Carrer de Maria Aurèlia Capmany, 40, E-17003, Girona, Spain. E-mail:
| | - Margarita Martinez-Medina
- Laboratori de Microbiologia Molecular, Departament de Biologia, Universitat de Girona, Girona, Spain
| |
Collapse
|
130
|
Mirza A, Mao-Draayer Y. The gut microbiome and microbial translocation in multiple sclerosis. Clin Immunol 2017; 183:213-224. [PMID: 28286112 DOI: 10.1016/j.clim.2017.03.001] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 02/03/2017] [Accepted: 03/07/2017] [Indexed: 02/07/2023]
Abstract
Individuals with multiple sclerosis (MS) have a distinct intestinal microbial community (microbiota) and increased low-grade translocation of bacteria from the intestines into the circulation. The observed change of intestinal bacteria in MS patients regulate immune functions involved in MS pathogenesis. These functions include: systemic and central nervous system (CNS) immunity (including peripheral regulatory T cell function), the blood-brain barrier (BBB) permeability and CNS-resident cell activity. This review discusses the MS intestinal microbiota implication on MS systemic- and CNS-immunopathology. We introduce the possible contributions of MS low-grade microbial translocation (LG-MT) to the development of MS, and end on a discussion on microbiota therapies for MS patients.
Collapse
Affiliation(s)
- Ali Mirza
- Department of Microbiology and Immunology, University of Michigan School of Medicine, 4258 Alfred Taubman Biomedical Sciences Research Bldg. 109 Zina Pitcher Place, Ann Arbor, MI 48109-2200, United States; Department of Neurology, University of Michigan School of Medicine, 4258 Alfred Taubman Biomedical Sciences Research Bldg. 109 Zina Pitcher Place, Ann Arbor, MI 48109-2200, United States
| | - Yang Mao-Draayer
- Department of Neurology, University of Michigan School of Medicine, 4015 Alfred Taubman Biomedical Sciences Research Bldg. 109 Zina Pitcher Place, Ann Arbor, MI 48109-2200, United States.
| |
Collapse
|
131
|
Breyner NM, Michon C, de Sousa CS, Vilas Boas PB, Chain F, Azevedo VA, Langella P, Chatel JM. Microbial Anti-Inflammatory Molecule (MAM) from Faecalibacterium prausnitzii Shows a Protective Effect on DNBS and DSS-Induced Colitis Model in Mice through Inhibition of NF-κB Pathway. Front Microbiol 2017; 8:114. [PMID: 28203226 PMCID: PMC5285381 DOI: 10.3389/fmicb.2017.00114] [Citation(s) in RCA: 127] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 01/17/2017] [Indexed: 12/19/2022] Open
Abstract
Faecalibacterium prausnitzii and its supernatant showed protective effects in different chemically-induced colitis models in mice. Recently, we described 7 peptides found in the F. prausnitzii supernatant, all belonging to a protein called Microbial Anti-inflammatory Molecule (MAM). These peptides were able to inhibit NF-κB pathway in vitro and showed anti-inflammatory properties in vivo in a DiNitroBenzene Sulfate (DNBS)-induced colitis model. In this current proof we tested MAM effect on NF-κB pathway in vivo, using a transgenic model of mice producing luciferase under the control of NF-κB promoter. Moreover, we tested this protein on Dextran Sodium Sulfate (DSS)-induced colitis in mice. To study the effect of MAM we orally administered to the mice a Lactococcus lactis strain carrying a plasmid containing the cDNA of MAM under the control of a eukaryotic promoter. L. lactis delivered plasmids in epithelial cells of the intestinal membrane allowing thus the production of MAM directly by host. We showed that MAM administration inhibits NF-κB pathway in vivo. We confirmed the anti-inflammatory properties of MAM in DNBS-induced colitis but also in DSS model. In DSS model MAM was able to inhibit Th1 and Th17 immune response while in DNBS model MAM reduced Th1, Th2, and Th17 immune response and increased TGFβ production.
Collapse
Affiliation(s)
- Natalia M Breyner
- Micalis Institute, Institut National de la Recherche Agronomique (INRA), AgroParisTech, Université Paris-Saclay Jouy-en-Josas, France
| | - Cristophe Michon
- Micalis Institute, Institut National de la Recherche Agronomique (INRA), AgroParisTech, Université Paris-Saclay Jouy-en-Josas, France
| | - Cassiana S de Sousa
- Micalis Institute, Institut National de la Recherche Agronomique (INRA), AgroParisTech, Université Paris-SaclayJouy-en-Josas, France; Laboratorio de Genetica Celular e Molecular, Departamento de Microbiologia, Universidade Federal de Minas GeraisBelo Horizonte, Brazil
| | - Priscilla B Vilas Boas
- Micalis Institute, Institut National de la Recherche Agronomique (INRA), AgroParisTech, Université Paris-SaclayJouy-en-Josas, France; Laboratorio de Genetica Celular e Molecular, Departamento de Microbiologia, Universidade Federal de Minas GeraisBelo Horizonte, Brazil
| | - Florian Chain
- Micalis Institute, Institut National de la Recherche Agronomique (INRA), AgroParisTech, Université Paris-Saclay Jouy-en-Josas, France
| | - Vasco A Azevedo
- Laboratorio de Genetica Celular e Molecular, Departamento de Microbiologia, Universidade Federal de Minas Gerais Belo Horizonte, Brazil
| | - Philippe Langella
- Micalis Institute, Institut National de la Recherche Agronomique (INRA), AgroParisTech, Université Paris-Saclay Jouy-en-Josas, France
| | - Jean M Chatel
- Micalis Institute, Institut National de la Recherche Agronomique (INRA), AgroParisTech, Université Paris-Saclay Jouy-en-Josas, France
| |
Collapse
|
132
|
De Weirdt R, Hernandez-Sanabria E, Fievez V, Mees E, Geirnaert A, Van Herreweghen F, Vilchez-Vargas R, Van den Abbeele P, Jauregui R, Pieper DH, Vlaeminck B, Van de Wiele T. Mucosa-associated biohydrogenating microbes protect the simulated colon microbiome from stress associated with high concentrations of poly-unsaturated fat. Environ Microbiol 2017; 19:722-739. [DOI: 10.1111/1462-2920.13622] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Rosemarie De Weirdt
- Center for Microbial Ecology and Technology (CMET), Ghent University; Coupure Links 653 Ghent BE-9000 Belgium
| | - Emma Hernandez-Sanabria
- Center for Microbial Ecology and Technology (CMET), Ghent University; Coupure Links 653 Ghent BE-9000 Belgium
| | - Veerle Fievez
- Laboratory for Animal Nutrition and Product Quality (Lanupro); Ghent University; Proefhoevestraat 10 Melle BE-9090 Belgium
| | - Eva Mees
- Center for Microbial Ecology and Technology (CMET), Ghent University; Coupure Links 653 Ghent BE-9000 Belgium
| | - Annelies Geirnaert
- Center for Microbial Ecology and Technology (CMET), Ghent University; Coupure Links 653 Ghent BE-9000 Belgium
| | - Florence Van Herreweghen
- Center for Microbial Ecology and Technology (CMET), Ghent University; Coupure Links 653 Ghent BE-9000 Belgium
| | - Ramiro Vilchez-Vargas
- Center for Microbial Ecology and Technology (CMET), Ghent University; Coupure Links 653 Ghent BE-9000 Belgium
| | - Pieter Van den Abbeele
- Center for Microbial Ecology and Technology (CMET), Ghent University; Coupure Links 653 Ghent BE-9000 Belgium
| | - Ruy Jauregui
- Microbial Interactions and Processes Research Group, Department of Molecular Infection Biology; Helmholtz Centre for Infection Research; Inhoffenstraβe 7 Braunschweig D-38124 Germany
| | - Dietmar H. Pieper
- Microbial Interactions and Processes Research Group, Department of Molecular Infection Biology; Helmholtz Centre for Infection Research; Inhoffenstraβe 7 Braunschweig D-38124 Germany
| | - Bruno Vlaeminck
- Laboratory for Animal Nutrition and Product Quality (Lanupro); Ghent University; Proefhoevestraat 10 Melle BE-9090 Belgium
| | - Tom Van de Wiele
- Center for Microbial Ecology and Technology (CMET), Ghent University; Coupure Links 653 Ghent BE-9000 Belgium
| |
Collapse
|
133
|
Alvarez CS, Badia J, Bosch M, Giménez R, Baldomà L. Outer Membrane Vesicles and Soluble Factors Released by Probiotic Escherichia coli Nissle 1917 and Commensal ECOR63 Enhance Barrier Function by Regulating Expression of Tight Junction Proteins in Intestinal Epithelial Cells. Front Microbiol 2016; 7:1981. [PMID: 28018313 PMCID: PMC5156689 DOI: 10.3389/fmicb.2016.01981] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 11/25/2016] [Indexed: 12/16/2022] Open
Abstract
The gastrointestinal epithelial layer forms a physical and biochemical barrier that maintains the segregation between host and intestinal microbiota. The integrity of this barrier is critical in maintaining homeostasis in the body and its dysfunction is linked to a variety of illnesses, especially inflammatory bowel disease. Gut microbes, and particularly probiotic bacteria, modulate the barrier integrity by reducing gut permeability and reinforcing tight junctions. Probiotic Escherichia coli Nissle 1917 (EcN) is a good colonizer of the human gut with proven therapeutic efficacy in the remission of ulcerative colitis in humans. EcN positively modulates the intestinal epithelial barrier through upregulation and redistribution of the tight junction proteins ZO-1, ZO-2 and claudin-14. Upregulation of claudin-14 has been attributed to the secreted protein TcpC. Whether regulation of ZO-1 and ZO-2 is mediated by EcN secreted factors remains unknown. The aim of this study was to explore whether outer membrane vesicles (OMVs) released by EcN strengthen the epithelial barrier. This study includes other E. coli strains of human intestinal origin that contain the tcpC gene, such as ECOR63. Cell-free supernatants collected from the wild-type strains and from the derived tcpC mutants were fractionated into isolated OMVs and soluble secreted factors. The impact of these extracellular fractions on the epithelial barrier was evaluated by measuring transepithelial resistance and expression of several tight junction proteins in T-84 and Caco-2 polarized monolayers. Our results show that the strengthening activity of EcN and ECOR63 does not exclusively depend on TcpC. Both OMVs and soluble factors secreted by these strains promote upregulation of ZO-1 and claudin-14, and down-regulation of claudin-2. The OMVs-mediated effects are TcpC-independent. Soluble secreted TcpC contributes to the upregulation of ZO-1 and claudin-14, but this protein has no effect on the transcriptional regulation of claudin-2. Thus, in addition to OMVs and TcpC, other active factors released by these microbiota strains contribute to the reinforcement of the epithelial barrier.
Collapse
Affiliation(s)
- Carina-Shianya Alvarez
- Secció de Bioquímica i Biologia Molecular, Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de BarcelonaBarcelona, Spain; Institut de Biomedicina de la Universitat de Barcelona, Institut de Recerca Sant Joan De DéuBarcelona, Spain
| | - Josefa Badia
- Secció de Bioquímica i Biologia Molecular, Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de BarcelonaBarcelona, Spain; Institut de Biomedicina de la Universitat de Barcelona, Institut de Recerca Sant Joan De DéuBarcelona, Spain
| | - Manel Bosch
- Unitat de Microscòpia Òptica Avançada, Centres Científics i Tecnològics, Universitat de Barcelona Barcelona, Spain
| | - Rosa Giménez
- Secció de Bioquímica i Biologia Molecular, Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de BarcelonaBarcelona, Spain; Institut de Biomedicina de la Universitat de Barcelona, Institut de Recerca Sant Joan De DéuBarcelona, Spain
| | - Laura Baldomà
- Secció de Bioquímica i Biologia Molecular, Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de BarcelonaBarcelona, Spain; Institut de Biomedicina de la Universitat de Barcelona, Institut de Recerca Sant Joan De DéuBarcelona, Spain
| |
Collapse
|
134
|
Protective Role of Postbiotic Mediators Secreted by Lactobacillus rhamnosus GG Versus Lipopolysaccharide-induced Damage in Human Colonic Smooth Muscle Cells. J Clin Gastroenterol 2016; 50 Suppl 2, Proceedings from the 8th Probiotics, Prebiotics & New Foods for Microbiota and Human Health meeting held in Rome, Italy on September 13-15, 2015:S140-S144. [PMID: 27741159 DOI: 10.1097/mcg.0000000000000681] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Some beneficial effects of probiotics may be due to secreted probiotic-derived factors, identified as "postbiotic" mediators. The aim of this study was to evaluate whether supernatants harvested from Lactobacillus rhamnosus GG (LGG) cultures (ATCC53103 strain) protect colonic human smooth muscle cells (HSMCs) from lipopolysaccharide (LPS)-induced myogenic damage. MATERIALS AND METHODS LGG was grown in de Man, Rogosa, Share medium at 37°C and samples were collected in middle and late exponential, stationary, and overnight phases. Supernatants were recovered by centrifugation, filtered, and stored at -20°C. The primary HSMCs culture was exposed for 24 hours to purified LPS of a pathogen strain of Escherichia coli (O111:B4) (1 μg/mL) with and without supernatants. Postbiotic effects were evaluated on the basis of HSMCs morphofunctional alterations and interleukin-6 (IL-6) production. Data are expressed as mean±SE (P<0.05 significant). RESULTS LPS induced persistent, significant, 20.5%±0.7% cell shortening and 34.5%±2.2% decrease in acetylcholine-induced contraction of human HSMCs. These morphofunctional alterations were paralleled to a 365.65%±203.13% increase in IL-6 production. All these effects were dose-dependently reduced by LGG supernatants. Supernatants of the middle exponential phase already partially restored LPS-induced cell shortening by 57.34%±12.7% and IL-6 increase by 145.8%±4.3% but had no effect on LPS-induced inhibition of contraction. Maximal protective effects were obtained with supernatants of the late stationary phase with LPS-induced cell shortening restored by 84.1%±4.7%, inhibition of contraction by 85.5%±6.4%, and IL-6 basal production by 92.7%±1.2%. CONCLUSIONS LGG-derived products are able to protect human SMCs from LPS-induced myogenic damage. Novel insights have been provided for the possibility that LGG-derived products could reduce the risk of progression to postinfective motor disorders.
Collapse
|
135
|
Bribi N, Algieri F, Rodriguez-Nogales A, Vezza T, Garrido-Mesa J, Utrilla MP, Del Mar Contreras M, Maiza F, Segura-Carretero A, Rodriguez-Cabezas ME, Gálvez J. Intestinal anti-inflammatory effects of total alkaloid extract from Fumaria capreolata in the DNBS model of mice colitis and intestinal epithelial CMT93 cells. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2016; 23:901-13. [PMID: 27387398 DOI: 10.1016/j.phymed.2016.05.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 05/04/2016] [Accepted: 05/10/2016] [Indexed: 05/27/2023]
Abstract
BACKGROUND Fumaria capreolata L. (Papaveraceae) is a botanical drug used in North Africa for its gastro-intestinal and anti-inflammatory properties. It is characterized for the presence of several alkaloids that could be responsible for some of its effects, including an immunomodulatory activity. PURPOSE To test in vivo the intestinal anti-inflammatory properties of the total alkaloid fraction extracted from the aerial parts of F. capreolata (AFC), and to evaluate its effects on an intestinal epithelial cell line. STUDY DESIGN AND METHODS AFC was chemically characterized by liquid chromatography coupled to diode array detection and high resolution mass spectrometry. Different doses of AFC (25, 50 and 100mg/kg) were assayed in the DNBS model of experimental colitis in mice, and the colonic damage was evaluated both histologically and biochemically. In addition, in vitro experiments were performed with this alkaloid fraction on the mouse intestinal epithelial cell line CMT93 stimulated with LPS. RESULTS The chemical analysis of AFC revealed the presence of 23 alkaloids, being the most abundants stylopine, protopine and coptisine. Oral administration of AFC produced a significant inhibition of the release and the expression of IL-6 and TNF-α in the colonic tissue. It also suppressed in vivo the transcription of other pro-inflammatory mediators such as IL-1β, iNOS, IL-12 and IL-17. Furthermore, AFC showed an immunomodulatory effect in vitro since it was able to inhibit the mRNA expression of IL-6, TNF-α and ICAM-1. Moreover, the beneficial effect of AFC in the colitic mice could also be associated with the normalization of the expression of MUC-2 and ZO-1, which are important for the intestinal epithelial integrity. CONCLUSION The present study suggests that AFC, containing 1.3% of stylopine and 0.9% of protopine, significantly exerted intestinal anti-inflammatory effects in an experimental model of mouse colitis. This fact could be related to a modulation of the intestinal immune response and a restoration of the intestinal epithelial function.
Collapse
Affiliation(s)
- Noureddine Bribi
- Laboratoire de Biotechnologies Végétales et Ethnobotanique, Faculté des Sciences de la Nature et de la Vie, Université de Bejaia, 06000 Bejaia, Algeria; CIBER-EHD, Department of Pharmacology, ibs.GRANADA, Center for Biomedical Research (CIBM), University of Granada, Avenida del Conocimiento s/n 18016-Armilla, Granada, Spain
| | - Francesca Algieri
- CIBER-EHD, Department of Pharmacology, ibs.GRANADA, Center for Biomedical Research (CIBM), University of Granada, Avenida del Conocimiento s/n 18016-Armilla, Granada, Spain
| | - Alba Rodriguez-Nogales
- CIBER-EHD, Department of Pharmacology, ibs.GRANADA, Center for Biomedical Research (CIBM), University of Granada, Avenida del Conocimiento s/n 18016-Armilla, Granada, Spain
| | - Teresa Vezza
- CIBER-EHD, Department of Pharmacology, ibs.GRANADA, Center for Biomedical Research (CIBM), University of Granada, Avenida del Conocimiento s/n 18016-Armilla, Granada, Spain
| | - Jose Garrido-Mesa
- CIBER-EHD, Department of Pharmacology, ibs.GRANADA, Center for Biomedical Research (CIBM), University of Granada, Avenida del Conocimiento s/n 18016-Armilla, Granada, Spain
| | - María Pilar Utrilla
- CIBER-EHD, Department of Pharmacology, ibs.GRANADA, Center for Biomedical Research (CIBM), University of Granada, Avenida del Conocimiento s/n 18016-Armilla, Granada, Spain
| | - María Del Mar Contreras
- Department of Analytical Chemistry, Faculty of Sciences, University of Granada, Avenida Fuentenueva s/n, 18071-Granada, Spain; Research and Development Centre for Functional Food (CIDAF), Health-Science Technological Park, Avenida del Conocimiento 37, 18016-Granada, Spain
| | - Fadila Maiza
- Laboratoire de Biotechnologies Végétales et Ethnobotanique, Faculté des Sciences de la Nature et de la Vie, Université de Bejaia, 06000 Bejaia, Algeria
| | - Antonio Segura-Carretero
- Department of Analytical Chemistry, Faculty of Sciences, University of Granada, Avenida Fuentenueva s/n, 18071-Granada, Spain; Research and Development Centre for Functional Food (CIDAF), Health-Science Technological Park, Avenida del Conocimiento 37, 18016-Granada, Spain
| | - Maria Elena Rodriguez-Cabezas
- CIBER-EHD, Department of Pharmacology, ibs.GRANADA, Center for Biomedical Research (CIBM), University of Granada, Avenida del Conocimiento s/n 18016-Armilla, Granada, Spain
| | - Julio Gálvez
- CIBER-EHD, Department of Pharmacology, ibs.GRANADA, Center for Biomedical Research (CIBM), University of Granada, Avenida del Conocimiento s/n 18016-Armilla, Granada, Spain.
| |
Collapse
|
136
|
Dolan KT, Chang EB. Diet, gut microbes, and the pathogenesis of inflammatory bowel diseases. Mol Nutr Food Res 2016; 61. [PMID: 27346644 DOI: 10.1002/mnfr.201600129] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Revised: 06/09/2016] [Accepted: 06/15/2016] [Indexed: 12/14/2022]
Abstract
The rising incidence of inflammatory bowel diseases in recent decades has notably paralleled changing lifestyle habits in Western nations, which are now making their way into more traditional societies. Diet plays a key role in IBD pathogenesis, and there is a growing appreciation that the interaction between diet and microbes in a susceptible person contributes significantly to the onset of disease. In this review, we examine what is known about dietary and microbial factors that promote IBD. We summarize recent findings regarding the effects of diet in IBD epidemiology from prospective population cohort studies, as well as new insights into IBD-associated dysbiosis. Microbial metabolism of dietary components can influence the epithelial barrier and the mucosal immune system, and understanding how these interactions generate or suppress inflammation will be a significant focus of IBD research. Our knowledge of dietary and microbial risk factors for IBD provides important considerations for developing therapeutic approaches through dietary modification or re-shaping the microbiota. We conclude by calling for increased sophistication in designing studies on the role of diet and microbes in IBD pathogenesis and disease resolution in order to accelerate progress in response to the growing challenge posed by these complex disorders.
Collapse
Affiliation(s)
- Kyle T Dolan
- Section of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, The University of Chicago
| | - Eugene B Chang
- Section of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, The University of Chicago
| |
Collapse
|
137
|
Magnusson MK, Strid H, Sapnara M, Lasson A, Bajor A, Ung KA, Öhman L. Anti-TNF Therapy Response in Patients with Ulcerative Colitis Is Associated with Colonic Antimicrobial Peptide Expression and Microbiota Composition. J Crohns Colitis 2016; 10:943-52. [PMID: 26896085 DOI: 10.1093/ecco-jcc/jjw051] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 02/16/2016] [Indexed: 02/08/2023]
Abstract
BACKGROUND AND AIMS Anti-tumour necrosis factor [TNF] therapy is used in patients with ulcerative colitis [UC], but not all patients respond to treatment. Antimicrobial peptides [AMPs] and the gut microbiota are essential for gut homeostasis and may be important for treatment outcome. The aim of this study was to determine AMP and microbiota profiles in patients with UC before anti-TNF therapy start and correlate these data to treatment outcome. METHODS Serum and biopsies were obtained from UC patients naïve to biological therapy [n = 56] before anti-TNF therapy start [baseline]. Fecal samples were taken at baseline and Weeks 2 and 6. Quantitative proteomic analysis was performed in mucosal biopsies. Expression of AMPs and cytokines was determined in biopsies and serum. Microbiota analysis of fecal samples was performed using GA-map™ Dysbiosis Test and real-time quantitative polymerase chain reaction [rtPCR]. Treatment response was evaluated 12-14 weeks after baseline. RESULTS At baseline, proteomic analysis of biopsies showed that treatment responders and non-responders had differential expression of AMPs. Eleven AMP and AMP-related genes were analysed by rtPCR in mucosal biopsies and could together discriminate responders from non-responders at baseline. The most important nominators for response were increased expression of defensin 5 and eosinophilic cationic protein. Microbiota analysis revealed lower dysbiosis indexes and higher abundance of Faecalibacterium prausnitzii in responders compared with non-responders at baseline. Also, abundance of F. prausnitzii increased during induction therapy in responders. CONCLUSIONS Anti-TNF therapy responders and non-responders display distinctly separate patterns of mucosal AMP expression and gut microbiota before treatment start. This indicates that intestinal antimicrobial/microbial composition can influence treatment outcome.
Collapse
Affiliation(s)
- Maria K Magnusson
- Department of Microbiology and Immunology, University of Gothenburg, Institute for Biomedicine, Gothenburg, Sweden Department of Internal Medicine and Clinical Nutrition, University of Gothenburg, Institute for Medicine, Gothenburg, Sweden
| | - Hans Strid
- Department of Internal Medicine and Clinical Nutrition, University of Gothenburg, Institute for Medicine, Gothenburg, Sweden Södra Älvsborg Hospital, Department of Internal Medicine, Borås, Sweden
| | - Maria Sapnara
- Department of Microbiology and Immunology, University of Gothenburg, Institute for Biomedicine, Gothenburg, Sweden Department of Internal Medicine and Clinical Nutrition, University of Gothenburg, Institute for Medicine, Gothenburg, Sweden
| | - Anders Lasson
- Södra Älvsborg Hospital, Department of Internal Medicine, Borås, Sweden
| | - Antal Bajor
- Department of Internal Medicine and Clinical Nutrition, University of Gothenburg, Institute for Medicine, Gothenburg, Sweden Södra Älvsborg Hospital, Department of Internal Medicine, Borås, Sweden
| | - Kjell-Arne Ung
- Kärnsjukhuset, Department of Internal Medicine, Skövde, Sweden
| | - Lena Öhman
- Department of Microbiology and Immunology, University of Gothenburg, Institute for Biomedicine, Gothenburg, Sweden Department of Internal Medicine and Clinical Nutrition, University of Gothenburg, Institute for Medicine, Gothenburg, Sweden School of Health and Education, University of Skövde, Skövde, Sweden
| |
Collapse
|
138
|
Hansen AK, Krych Ł, Nielsen DS, Hansen CHF. A Review of Applied Aspects of Dealing with Gut Microbiota Impact on Rodent Models. ILAR J 2016; 56:250-64. [PMID: 26323634 DOI: 10.1093/ilar/ilv010] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The gut microbiota (GM) affects numerous human diseases, as well as rodent models for these. We will review this impact and summarize ways to handle this challenge in animal research. The GM is complex, with the largest fractions being the gram-positive phylum Firmicutes and the gram-negative phylum Bacteroidetes. Other important phyla are the gram-negative phyla Proteobacteria and Verrucomicrobia, and the gram-positive phylum Actinobacteria. GM members influence models for diseases, such as inflammatory bowel diseases, allergies, autoimmunity, cancer, and neuropsychiatric diseases. GM characterization of all individual animals and incorporation of their GM composition in data evaluation may therefore be considered in future protocols. Germfree isolator-housed rodents or rodents made virtually germ free by antibiotic cocktails can be used to study diverse microbial influences on disease expression. Through subsequent inoculation with selected strains or cocktails of microbes, new "defined flora" models can yield valuable knowledge on the impact of the GM, and of specific GM members and their interactions, on important disease phenotypes and mechanisms. Rodent husbandry and microbial quality assurance practices will be important to ensure and confirm appropriate and research relevant GM.
Collapse
Affiliation(s)
- Axel Kornerup Hansen
- Axel Kornerup Hansen, DVM, DVsc, DipECLAM, Professor, Section of Experimental Animal Models, Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Thorvaldsensvej 57, 1871 Frederiksberg C, Denmark. Łukasz Krych, MSc, PhD, Postdoc, Department of Food Science, Faculty of Science, University of Copenhagen, Rolighedsvej 26, 1958 Frederiksberg C, Denmark. Dennis Sandris Nielsen, MSc, PhD, Associate Professor, Department of Food Science, Faculty of Science, University of Copenhagen, Rolighedsvej 26, 1958 Frederiksberg C, Denmark. Camilla Hartmann Friis Hansen, DVM, PhD, Assistant Professor, Section of Experimental Animal Models, Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Thorvaldsensvej 57, 1871 Frederiksberg C, Denmark
| | - Łukasz Krych
- Axel Kornerup Hansen, DVM, DVsc, DipECLAM, Professor, Section of Experimental Animal Models, Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Thorvaldsensvej 57, 1871 Frederiksberg C, Denmark. Łukasz Krych, MSc, PhD, Postdoc, Department of Food Science, Faculty of Science, University of Copenhagen, Rolighedsvej 26, 1958 Frederiksberg C, Denmark. Dennis Sandris Nielsen, MSc, PhD, Associate Professor, Department of Food Science, Faculty of Science, University of Copenhagen, Rolighedsvej 26, 1958 Frederiksberg C, Denmark. Camilla Hartmann Friis Hansen, DVM, PhD, Assistant Professor, Section of Experimental Animal Models, Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Thorvaldsensvej 57, 1871 Frederiksberg C, Denmark
| | - Dennis Sandris Nielsen
- Axel Kornerup Hansen, DVM, DVsc, DipECLAM, Professor, Section of Experimental Animal Models, Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Thorvaldsensvej 57, 1871 Frederiksberg C, Denmark. Łukasz Krych, MSc, PhD, Postdoc, Department of Food Science, Faculty of Science, University of Copenhagen, Rolighedsvej 26, 1958 Frederiksberg C, Denmark. Dennis Sandris Nielsen, MSc, PhD, Associate Professor, Department of Food Science, Faculty of Science, University of Copenhagen, Rolighedsvej 26, 1958 Frederiksberg C, Denmark. Camilla Hartmann Friis Hansen, DVM, PhD, Assistant Professor, Section of Experimental Animal Models, Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Thorvaldsensvej 57, 1871 Frederiksberg C, Denmark
| | - Camilla Hartmann Friis Hansen
- Axel Kornerup Hansen, DVM, DVsc, DipECLAM, Professor, Section of Experimental Animal Models, Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Thorvaldsensvej 57, 1871 Frederiksberg C, Denmark. Łukasz Krych, MSc, PhD, Postdoc, Department of Food Science, Faculty of Science, University of Copenhagen, Rolighedsvej 26, 1958 Frederiksberg C, Denmark. Dennis Sandris Nielsen, MSc, PhD, Associate Professor, Department of Food Science, Faculty of Science, University of Copenhagen, Rolighedsvej 26, 1958 Frederiksberg C, Denmark. Camilla Hartmann Friis Hansen, DVM, PhD, Assistant Professor, Section of Experimental Animal Models, Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Thorvaldsensvej 57, 1871 Frederiksberg C, Denmark
| |
Collapse
|
139
|
The Microbiota Determines Susceptibility to Experimental Autoimmune Uveoretinitis. J Immunol Res 2016; 2016:5065703. [PMID: 27294159 PMCID: PMC4886056 DOI: 10.1155/2016/5065703] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 03/08/2016] [Accepted: 04/11/2016] [Indexed: 02/07/2023] Open
Abstract
The microbiota is a crucial modulator of the immune system. Here, we evaluated how its absence or reduction modifies the inflammatory response in the murine model of experimental autoimmune uveoretinitis (EAU). We induced EAU in germ-free (GF) or conventionally housed (CV) mice and in CV mice treated with a combination of broad-spectrum antibiotics either from the day of EAU induction or from one week prior to induction of disease. The severity of the inflammation was assessed by fundus biomicroscopy or by histology, including immunohistology. The immunophenotyping of T cells in local and distant lymph nodes was performed by flow cytometry. We found that GF mice and mice where the microbiota was reduced one week before EAU induction were protected from severe autoimmune inflammation. GF mice had lower numbers of infiltrating macrophages and significantly less T cell infiltration in the retina than CV mice with EAU. GF mice also had reduced numbers of IFN-γ and IL-17-producing T cells and increased numbers of regulatory T cells in the eye-draining lymph nodes. These data suggest that the presence of microbiota during autoantigen recognition regulates the inflammatory response by influencing the adaptive immune response.
Collapse
|
140
|
Martín R, Laval L, Chain F, Miquel S, Natividad J, Cherbuy C, Sokol H, Verdu EF, van Hylckama Vlieg J, Bermudez-Humaran LG, Smokvina T, Langella P. Bifidobacterium animalis ssp. lactis CNCM-I2494 Restores Gut Barrier Permeability in Chronically Low-Grade Inflamed Mice. Front Microbiol 2016; 7:608. [PMID: 27199937 PMCID: PMC4858658 DOI: 10.3389/fmicb.2016.00608] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 04/12/2016] [Indexed: 01/15/2023] Open
Abstract
Growing evidence supports the efficacy of many probiotic strains in the management of gastrointestinal disorders associated with deregulated intestinal barrier function and/or structure. In particular, bifidobacteria have been studied for their efficacy to both prevent and treat a broad spectrum of animal and/or human gut disorders. The aim of the current work was thus to evaluate effects on intestinal barrier function of Bifidobacterium animalis ssp. lactis CNCM-I2494, a strain used in fermented dairy products. A chronic dinitrobenzene sulfonic acid (DNBS)-induced low-grade inflammation model causing gut dysfunction in mice was used in order to study markers of inflammation, intestinal permeability, and immune function in the presence of the bacterial strain. In this chronic low-grade inflammation mice model several parameters pointed out the absence of an over active inflammation process. However, gut permeability, lymphocyte populations, and colonic cytokines were found to be altered. B. animalis ssp. lactis CNCM-I2494 was able to protect barrier functions by restoring intestinal permeability, colonic goblet cell populations, and cytokine levels. Furthermore, tight junction (TJ) proteins levels were also measured by qRT-PCR showing the ability of this strain to specifically normalize the level of several TJ proteins, in particular for claudin-4. Finally, B. lactis strain counterbalanced CD4+ lymphocyte alterations in both spleen and mesenteric lymphoid nodes. It restores the Th1/Th2 ratio altered by the DNBS challenge (which locally augments CD4+ Th1 cells) by increasing the Th2 response as measured by the increase in the production of major representative Th2 cytokines (IL-4, IL-5, and IL-10). Altogether, these data suggest that B. animalis ssp. lactis CNCM-I2494 may efficiently prevent disorders associated with increased barrier permeability.
Collapse
Affiliation(s)
- Rebeca Martín
- Commensal and Probiotics-Host Interactions Laboratory, Micalis Institute, Institut National de la Recherche Agronomique, AgroParisTech, Université Paris-Saclay Jouy-en-Josas, France
| | - Laure Laval
- Commensal and Probiotics-Host Interactions Laboratory, Micalis Institute, Institut National de la Recherche Agronomique, AgroParisTech, Université Paris-SaclayJouy-en-Josas, France; Danone Nutricia ResearchPalaiseau, France
| | - Florian Chain
- Commensal and Probiotics-Host Interactions Laboratory, Micalis Institute, Institut National de la Recherche Agronomique, AgroParisTech, Université Paris-Saclay Jouy-en-Josas, France
| | - Sylvie Miquel
- Commensal and Probiotics-Host Interactions Laboratory, Micalis Institute, Institut National de la Recherche Agronomique, AgroParisTech, Université Paris-Saclay Jouy-en-Josas, France
| | - Jane Natividad
- Farncombe Family Digestive Health Research Institute, McMaster University Hamilton, ON, Canada
| | - Claire Cherbuy
- Commensal and Probiotics-Host Interactions Laboratory, Micalis Institute, Institut National de la Recherche Agronomique, AgroParisTech, Université Paris-Saclay Jouy-en-Josas, France
| | - Harry Sokol
- Commensal and Probiotics-Host Interactions Laboratory, Micalis Institute, Institut National de la Recherche Agronomique, AgroParisTech, Université Paris-SaclayJouy-en-Josas, France; ERL INSERM U 1057/UMR7203, Faculté de Médecine Saint-Antoine, Université Pierre et Marie CurieParis, France; Service de Gastroentérologie, Hôpital Saint-Antoine, Assistance Publique - Hôpitaux de ParisParis, France
| | - Elena F Verdu
- Farncombe Family Digestive Health Research Institute, McMaster University Hamilton, ON, Canada
| | | | - Luis G Bermudez-Humaran
- Commensal and Probiotics-Host Interactions Laboratory, Micalis Institute, Institut National de la Recherche Agronomique, AgroParisTech, Université Paris-Saclay Jouy-en-Josas, France
| | | | - Philippe Langella
- Commensal and Probiotics-Host Interactions Laboratory, Micalis Institute, Institut National de la Recherche Agronomique, AgroParisTech, Université Paris-Saclay Jouy-en-Josas, France
| |
Collapse
|
141
|
Quévrain E, Maubert MA, Michon C, Chain F, Marquant R, Tailhades J, Miquel S, Carlier L, Bermúdez-Humarán LG, Pigneur B, Lequin O, Kharrat P, Thomas G, Rainteau D, Aubry C, Breyner N, Afonso C, Lavielle S, Grill JP, Chassaing G, Chatel JM, Trugnan G, Xavier R, Langella P, Sokol H, Seksik P. Identification of an anti-inflammatory protein from Faecalibacterium prausnitzii, a commensal bacterium deficient in Crohn's disease. Gut 2016; 65:415-425. [PMID: 26045134 PMCID: PMC5136800 DOI: 10.1136/gutjnl-2014-307649] [Citation(s) in RCA: 511] [Impact Index Per Article: 63.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 05/21/2015] [Indexed: 12/13/2022]
Abstract
BACKGROUND Crohn's disease (CD)-associated dysbiosis is characterised by a loss of Faecalibacterium prausnitzii, whose culture supernatant exerts an anti-inflammatory effect both in vitro and in vivo. However, the chemical nature of the anti-inflammatory compounds has not yet been determined. METHODS Peptidomic analysis using mass spectrometry was applied to F. prausnitzii supernatant. Anti-inflammatory effects of identified peptides were tested in vitro directly on intestinal epithelial cell lines and on cell lines transfected with a plasmid construction coding for the candidate protein encompassing these peptides. In vivo, the cDNA of the candidate protein was delivered to the gut by recombinant lactic acid bacteria to prevent dinitrobenzene sulfonic acid (DNBS)-colitis in mice. RESULTS The seven peptides, identified in the F. prausnitzii culture supernatants, derived from a single microbial anti-inflammatory molecule (MAM), a protein of 15 kDa, and comprising 53% of non-polar residues. This last feature prevented the direct characterisation of the putative anti-inflammatory activity of MAM-derived peptides. Transfection of MAM cDNA in epithelial cells led to a significant decrease in the activation of the nuclear factor (NF)-κB pathway with a dose-dependent effect. Finally, the use of a food-grade bacterium, Lactococcus lactis, delivering a plasmid encoding MAM was able to alleviate DNBS-induced colitis in mice. CONCLUSIONS A 15 kDa protein with anti-inflammatory properties is produced by F. prausnitzii, a commensal bacterium involved in CD pathogenesis. This protein is able to inhibit the NF-κB pathway in intestinal epithelial cells and to prevent colitis in an animal model.
Collapse
Affiliation(s)
- E. Quévrain
- Sorbonne Universités, UPMC Univ Paris 06, LBM, 27 rue de Chaligny, F-75012, Paris, France.
,INSERM-ERL 1157 and Inflammation-Immunopathology-Biotherapy Department (DHU i2B), CHU Saint-Antoine 27 rue de Chaligny, F-75012 Paris, France.
,CNRS, UMR 7203 LBM, F-75005, Paris, France
| | - M. A. Maubert
- Sorbonne Universités, UPMC Univ Paris 06, LBM, 27 rue de Chaligny, F-75012, Paris, France.
,INSERM-ERL 1157 and Inflammation-Immunopathology-Biotherapy Department (DHU i2B), CHU Saint-Antoine 27 rue de Chaligny, F-75012 Paris, France.
,CNRS, UMR 7203 LBM, F-75005, Paris, France
,APHP, Hôpital Saint Antoine - Département PM2 Plateforme de Métabolomique, Peptidomique et dosage de Médicaments, F-75012 Paris, France
| | - C. Michon
- INRA, UMR1319 Micalis, F-78350 Jouy-en-Josas, France.
,AgroParisTech, UMR Micalis, F-78350 Jouy-en-Josas, France
| | - F. Chain
- INRA, UMR1319 Micalis, F-78350 Jouy-en-Josas, France.
,AgroParisTech, UMR Micalis, F-78350 Jouy-en-Josas, France
| | - R. Marquant
- Sorbonne Universités, UPMC Univ Paris 06, LBM, 27 rue de Chaligny, F-75012, Paris, France.
,CNRS, UMR 7203 LBM, F-75005, Paris, France
,Ecole Normale Supérieure- PSL Research University, Département de Chimie 24 rue Lhomond, F-75005 Paris, France
| | - J. Tailhades
- Sorbonne Universités, UPMC Univ Paris 06, LBM, 27 rue de Chaligny, F-75012, Paris, France.
,CNRS, UMR 7203 LBM, F-75005, Paris, France
,Ecole Normale Supérieure- PSL Research University, Département de Chimie 24 rue Lhomond, F-75005 Paris, France
| | - S. Miquel
- INRA, UMR1319 Micalis, F-78350 Jouy-en-Josas, France.
,AgroParisTech, UMR Micalis, F-78350 Jouy-en-Josas, France
| | - L. Carlier
- Sorbonne Universités, UPMC Univ Paris 06, LBM, 27 rue de Chaligny, F-75012, Paris, France.
,CNRS, UMR 7203 LBM, F-75005, Paris, France
,Ecole Normale Supérieure- PSL Research University, Département de Chimie 24 rue Lhomond, F-75005 Paris, France
| | - L. G. Bermúdez-Humarán
- INRA, UMR1319 Micalis, F-78350 Jouy-en-Josas, France.
,AgroParisTech, UMR Micalis, F-78350 Jouy-en-Josas, France
| | - B. Pigneur
- Sorbonne Universités, UPMC Univ Paris 06, LBM, 27 rue de Chaligny, F-75012, Paris, France.
,INSERM-ERL 1157 and Inflammation-Immunopathology-Biotherapy Department (DHU i2B), CHU Saint-Antoine 27 rue de Chaligny, F-75012 Paris, France.
,CNRS, UMR 7203 LBM, F-75005, Paris, France
| | - O. Lequin
- Sorbonne Universités, UPMC Univ Paris 06, LBM, 27 rue de Chaligny, F-75012, Paris, France.
,CNRS, UMR 7203 LBM, F-75005, Paris, France
,Ecole Normale Supérieure- PSL Research University, Département de Chimie 24 rue Lhomond, F-75005 Paris, France
| | - P. Kharrat
- INRA, UMR1319 Micalis, F-78350 Jouy-en-Josas, France.
,AgroParisTech, UMR Micalis, F-78350 Jouy-en-Josas, France
| | - G. Thomas
- Sorbonne Universités, UPMC Univ Paris 06, LBM, 27 rue de Chaligny, F-75012, Paris, France.
,INSERM-ERL 1157 and Inflammation-Immunopathology-Biotherapy Department (DHU i2B), CHU Saint-Antoine 27 rue de Chaligny, F-75012 Paris, France.
,CNRS, UMR 7203 LBM, F-75005, Paris, France
| | - D. Rainteau
- Sorbonne Universités, UPMC Univ Paris 06, LBM, 27 rue de Chaligny, F-75012, Paris, France.
,INSERM-ERL 1157 and Inflammation-Immunopathology-Biotherapy Department (DHU i2B), CHU Saint-Antoine 27 rue de Chaligny, F-75012 Paris, France.
,CNRS, UMR 7203 LBM, F-75005, Paris, France
,APHP, Hôpital Saint Antoine - Département PM2 Plateforme de Métabolomique, Peptidomique et dosage de Médicaments, F-75012 Paris, France
| | - C. Aubry
- INRA, UMR1319 Micalis, F-78350 Jouy-en-Josas, France.
,AgroParisTech, UMR Micalis, F-78350 Jouy-en-Josas, France
| | - N. Breyner
- INRA, UMR1319 Micalis, F-78350 Jouy-en-Josas, France.
,AgroParisTech, UMR Micalis, F-78350 Jouy-en-Josas, France
| | - C. Afonso
- Université de Rouen, UMR 6014 COBRA / IRCOF, F-76130 Mont Saint Aignan, France
| | - S. Lavielle
- Sorbonne Universités, UPMC Univ Paris 06, LBM, 27 rue de Chaligny, F-75012, Paris, France.
,CNRS, UMR 7203 LBM, F-75005, Paris, France
,Ecole Normale Supérieure- PSL Research University, Département de Chimie 24 rue Lhomond, F-75005 Paris, France
| | - J.-P. Grill
- Sorbonne Universités, UPMC Univ Paris 06, LBM, 27 rue de Chaligny, F-75012, Paris, France.
,INSERM-ERL 1157 and Inflammation-Immunopathology-Biotherapy Department (DHU i2B), CHU Saint-Antoine 27 rue de Chaligny, F-75012 Paris, France.
,CNRS, UMR 7203 LBM, F-75005, Paris, France
| | - G. Chassaing
- Sorbonne Universités, UPMC Univ Paris 06, LBM, 27 rue de Chaligny, F-75012, Paris, France.
,CNRS, UMR 7203 LBM, F-75005, Paris, France
,Ecole Normale Supérieure- PSL Research University, Département de Chimie 24 rue Lhomond, F-75005 Paris, France
| | - J. M. Chatel
- INRA, UMR1319 Micalis, F-78350 Jouy-en-Josas, France.
,AgroParisTech, UMR Micalis, F-78350 Jouy-en-Josas, France
| | - G. Trugnan
- Sorbonne Universités, UPMC Univ Paris 06, LBM, 27 rue de Chaligny, F-75012, Paris, France.
,INSERM-ERL 1157 and Inflammation-Immunopathology-Biotherapy Department (DHU i2B), CHU Saint-Antoine 27 rue de Chaligny, F-75012 Paris, France.
,CNRS, UMR 7203 LBM, F-75005, Paris, France
,APHP, Hôpital Saint Antoine - Département PM2 Plateforme de Métabolomique, Peptidomique et dosage de Médicaments, F-75012 Paris, France
| | - R. Xavier
- Center for Systems Biology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - P. Langella
- INRA, UMR1319 Micalis, F-78350 Jouy-en-Josas, France.
,AgroParisTech, UMR Micalis, F-78350 Jouy-en-Josas, France
| | - H. Sokol
- Sorbonne Universités, UPMC Univ Paris 06, LBM, 27 rue de Chaligny, F-75012, Paris, France.
,INSERM-ERL 1157 and Inflammation-Immunopathology-Biotherapy Department (DHU i2B), CHU Saint-Antoine 27 rue de Chaligny, F-75012 Paris, France.
,CNRS, UMR 7203 LBM, F-75005, Paris, France
,INRA, UMR1319 Micalis, F-78350 Jouy-en-Josas, France.
,APHP, Hôpital Saint Antoine – Service de Gastroentérologie et nutrition, F-75012 Paris, France
| | - P. Seksik
- Sorbonne Universités, UPMC Univ Paris 06, LBM, 27 rue de Chaligny, F-75012, Paris, France.
,INSERM-ERL 1157 and Inflammation-Immunopathology-Biotherapy Department (DHU i2B), CHU Saint-Antoine 27 rue de Chaligny, F-75012 Paris, France.
,CNRS, UMR 7203 LBM, F-75005, Paris, France
,APHP, Hôpital Saint Antoine – Service de Gastroentérologie et nutrition, F-75012 Paris, France
| |
Collapse
|
142
|
Hornef MW, Pabst O. Real friends: Faecalibacterium prausnitzii supports mucosal immune homeostasis. Gut 2016; 65:365-7. [PMID: 26531718 DOI: 10.1136/gutjnl-2015-310027] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 09/30/2015] [Indexed: 12/15/2022]
Affiliation(s)
- Mathias W Hornef
- Institute of Medical Microbiology, RWTH University Hospital, Aachen, Germany
| | - Oliver Pabst
- Institute of Molecular Medicine, RWTH University Hospital, Aachen, Germany
| |
Collapse
|
143
|
Patterson E, Ryan PM, Cryan JF, Dinan TG, Ross RP, Fitzgerald GF, Stanton C. Gut microbiota, obesity and diabetes. Postgrad Med J 2016; 92:286-300. [PMID: 26912499 DOI: 10.1136/postgradmedj-2015-133285] [Citation(s) in RCA: 328] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 01/28/2016] [Indexed: 02/06/2023]
Abstract
The central role of the intestinal microbiota in the progression and, equally, prevention of metabolic dysfunction is becoming abundantly apparent. The symbiotic relationship between intestinal microbiota and host ensures appropriate development of the metabolic system in humans. However, disturbances in composition and, in turn, functionality of the intestinal microbiota can disrupt gut barrier function, a trip switch for metabolic endotoxemia. This low-grade chronic inflammation, brought about by the influx of inflammatory bacterial fragments into circulation through a malfunctioning gut barrier, has considerable knock-on effects for host adiposity and insulin resistance. Conversely, recent evidence suggests that there are certain bacterial species that may interact with host metabolism through metabolite-mediated stimulation of enteric hormones and other systems outside of the gastrointestinal tract, such as the endocannabinoid system. When the abundance of these keystone species begins to decline, we see a collapse of the symbiosis, reflected in a deterioration of host metabolic health. This review will investigate the intricate axis between the microbiota and host metabolism, while also addressing the promising and novel field of probiotics as metabolic therapies.
Collapse
Affiliation(s)
- Elaine Patterson
- APC Microbiome Institute, University College Cork, Co. Cork, Ireland Food Biosciences Department, Teagasc Food Research Centre, Fermoy, Co. Cork, Ireland
| | - Paul M Ryan
- Food Biosciences Department, Teagasc Food Research Centre, Fermoy, Co. Cork, Ireland School of Microbiology, University College Cork, Co. Cork, Ireland
| | - John F Cryan
- APC Microbiome Institute, University College Cork, Co. Cork, Ireland Department of Anatomy and Neuroscience, University College Cork, Co. Cork, Ireland
| | - Timothy G Dinan
- APC Microbiome Institute, University College Cork, Co. Cork, Ireland Department of Psychiatry and Neurobehavioural Science, University College Cork, Co. Cork, Ireland
| | - R Paul Ross
- APC Microbiome Institute, University College Cork, Co. Cork, Ireland College of Science, Engineering and Food Science, University College Cork, Co. Cork, Ireland
| | - Gerald F Fitzgerald
- APC Microbiome Institute, University College Cork, Co. Cork, Ireland School of Microbiology, University College Cork, Co. Cork, Ireland
| | - Catherine Stanton
- APC Microbiome Institute, University College Cork, Co. Cork, Ireland Food Biosciences Department, Teagasc Food Research Centre, Fermoy, Co. Cork, Ireland
| |
Collapse
|
144
|
Abstract
This report summarises talks given at the 8th International Yakult Symposium, held on 23-24 April 2015 in Berlin. Two presentations explored different aspects of probiotic intervention: the small intestine as a probiotic target and inclusion of probiotics into integrative approaches to gastroenterology. Probiotic recommendations in gastroenterology guidelines and current data on probiotic efficacy in paediatric patients were reviewed. Updates were given on probiotic and gut microbiota research in obesity and obesity-related diseases, the gut-brain axis and development of psychobiotics, and the protective effects of equol-producing strains for prostate cancer. Recent studies were presented on probiotic benefit for antibiotic-associated diarrhoea and people with HIV, as well as protection against the adverse effects of a short-term high-fat diet. Aspects of probiotic mechanisms of activity were discussed, including immunomodulatory mechanisms and metabolite effects, the anti-inflammatory properties of Faecalibacterium prausnitzii, the relationship between periodontitis, microbial production of butyrate in the oral cavity and ageing, and the pathogenic mechanisms of Campylobacter. Finally, an insight was given on a recent expert meeting, which re-examined the probiotic definition, advised on the appropriate use and scope of the term and outlined different probiotic categories and the prevalence of different mechanisms of activity.
Collapse
|
145
|
Miquel S, Martín R, Lashermes A, Gillet M, Meleine M, Gelot A, Eschalier A, Ardid D, Bermúdez-Humarán LG, Sokol H, Thomas M, Theodorou V, Langella P, Carvalho FA. Anti-nociceptive effect of Faecalibacterium prausnitzii in non-inflammatory IBS-like models. Sci Rep 2016; 6:19399. [PMID: 26775847 PMCID: PMC4726104 DOI: 10.1038/srep19399] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 11/30/2015] [Indexed: 02/08/2023] Open
Abstract
Visceral pain and intestinal dysbiosis are associated with Irritable Bowel Syndrome (IBS), a common functional gastrointestinal disorder without available efficient therapies. In this study, a decrease of Faecalibacterium prausnitzii presence has been observed in an IBS-like rodent model induced by a neonatal maternal separation (NMS) stress. Moreover, it was investigated whether F. prausnitzii may have an impact on colonic sensitivity. The A2-165 reference strain, but not its supernatant, significantly decreased colonic hypersensitivity induced by either NMS in mice or partial restraint stress in rats. This effect was associated with a reinforcement of intestinal epithelial barrier. Thus, F. prausnitzii exhibits anti-nociceptive properties, indicating its potential to treat abdominal pain in IBS patients.
Collapse
Affiliation(s)
- S Miquel
- INRA, Commensal and Probiotics-Host Interactions Laboratory, UMR 1319 Micalis, F-78350 Jouy-en-Josas, France.,AgroParisTech, UMR1319 Micalis, F-78350 Jouy-en-Josas, France.,Laboratoire Microorganismes: Génome et Environnement, UMR CNRS 6023, 63000 Clermont-Ferrand, France.,Université d'Auvergne, 63000, Clermont-Ferrand, France
| | - R Martín
- INRA, Commensal and Probiotics-Host Interactions Laboratory, UMR 1319 Micalis, F-78350 Jouy-en-Josas, France.,AgroParisTech, UMR1319 Micalis, F-78350 Jouy-en-Josas, France
| | - A Lashermes
- Université d'Auvergne, 63000, Clermont-Ferrand, France.,INSERM 1107 Neuro-Dol, 630000 Clermont-Ferrand, France
| | - M Gillet
- Neuro-Gastroenterology and Nutrition Team, TOXALIM, UMR 1331-INRA/INP/UPS, F-31931 Toulouse, France
| | - M Meleine
- Université d'Auvergne, 63000, Clermont-Ferrand, France.,INSERM 1107 Neuro-Dol, 630000 Clermont-Ferrand, France
| | - A Gelot
- Université d'Auvergne, 63000, Clermont-Ferrand, France.,INSERM 1107 Neuro-Dol, 630000 Clermont-Ferrand, France
| | - A Eschalier
- Université d'Auvergne, 63000, Clermont-Ferrand, France.,INSERM 1107 Neuro-Dol, 630000 Clermont-Ferrand, France
| | - D Ardid
- Université d'Auvergne, 63000, Clermont-Ferrand, France.,INSERM 1107 Neuro-Dol, 630000 Clermont-Ferrand, France
| | - L G Bermúdez-Humarán
- INRA, Commensal and Probiotics-Host Interactions Laboratory, UMR 1319 Micalis, F-78350 Jouy-en-Josas, France.,AgroParisTech, UMR1319 Micalis, F-78350 Jouy-en-Josas, France
| | - H Sokol
- INRA, Commensal and Probiotics-Host Interactions Laboratory, UMR 1319 Micalis, F-78350 Jouy-en-Josas, France.,AgroParisTech, UMR1319 Micalis, F-78350 Jouy-en-Josas, France.,APHP, Hôpital Saint Antoine - Service de Gastroentérologie et nutrition, F-75012 Paris, France.,Sorbonne Universités, UPMC Univ Paris 06, LBM, 27 rue de Chaligny, F-75012 Paris, France.,INSERM-ERL 1157 and Inflammation-Immunopathology-Biotherapy Department (DHU i2B), CHU Saint-Antoine 27 rue de Chaligny, F-75012 Paris, France.,CNRS, UMR 7203 LBM, F-75005 Paris, France
| | - M Thomas
- INRA, Commensal and Probiotics-Host Interactions Laboratory, UMR 1319 Micalis, F-78350 Jouy-en-Josas, France.,AgroParisTech, UMR1319 Micalis, F-78350 Jouy-en-Josas, France
| | - V Theodorou
- Neuro-Gastroenterology and Nutrition Team, TOXALIM, UMR 1331-INRA/INP/UPS, F-31931 Toulouse, France
| | - P Langella
- INRA, Commensal and Probiotics-Host Interactions Laboratory, UMR 1319 Micalis, F-78350 Jouy-en-Josas, France.,AgroParisTech, UMR1319 Micalis, F-78350 Jouy-en-Josas, France
| | - F A Carvalho
- Université d'Auvergne, 63000, Clermont-Ferrand, France.,INSERM 1107 Neuro-Dol, 630000 Clermont-Ferrand, France
| |
Collapse
|
146
|
Rossi O, van Berkel LA, Chain F, Tanweer Khan M, Taverne N, Sokol H, Duncan SH, Flint HJ, Harmsen HJM, Langella P, Samsom JN, Wells JM. Faecalibacterium prausnitzii A2-165 has a high capacity to induce IL-10 in human and murine dendritic cells and modulates T cell responses. Sci Rep 2016; 6:18507. [PMID: 26725514 PMCID: PMC4698756 DOI: 10.1038/srep18507] [Citation(s) in RCA: 148] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 10/06/2015] [Indexed: 02/06/2023] Open
Abstract
Faecalibacterium prausnitzii strain A2-165 was previously reported to have anti-inflammatory properties and prevent colitis in a TNBS model. We compared the immunomodulatory properties of strain A2-165 to four different F. prausnitzii isolates and eight abundant intestinal commensals using human dendritic cells (DCs) and mouse BMDCs in vitro. Principal component analysis revealed that the cytokine response to F. prausnitzii A2-165 is distinct from the other strains in eliciting high amounts of IL-10 secretion. The mouse DNBS model of relapsing IBD was used to compare the protective effects of F. prausnitzii A2-165 and Clostridium hathewayi, a low secretor of IL-10, on the Th1-driven inflammatory response to DNBS; attenuation of disease parameters was only observed with F. prausnitzii. In an in vivo mouse model of nasal tolerance to ovalbumin, F. prausnitzii A2-165 enhanced ovalbumin-specific T cell proliferation and reduced the proportion of IFN-γ(+) T cells in CLNs. Similarly, in vitro F. prausnitzii A2-165 stimulated BMDCs increased ovalbumin-specific T cell proliferation and reduced the number of IFN-γ(+) T cells. These mechanisms may contribute to the anti-inflammatory effects of F. prausnitzii in colitis and support the notion that this abundant bacterium might contribute to immune homeostasis in the intestine via its anti-inflammatory properties.
Collapse
Affiliation(s)
- Oriana Rossi
- Host-Microbe Interactomics Group, University of Wageningen, 6708 WD, The Netherlands
| | - Lisette A van Berkel
- Department of Pediatrics, Erasmus Medical Center - Sophia Children's Hospital, 3015 CE, Rotterdam, The Netherlands
| | - Florian Chain
- Commensal and Probiotics-Host Interactions Laboratory, UMR 1319 Micalis, INRA, 78352, Jouy-en-Josas, France.,UMR 1319 Micalis, AgroparisTech, 78352, Jouy-en-Josas, France
| | - M Tanweer Khan
- Department of Medical Microbiology, University Medical Center Groningen, 9700 RB, Groningen, The Netherlands
| | - Nico Taverne
- Host-Microbe Interactomics Group, University of Wageningen, 6708 WD, The Netherlands
| | - Harry Sokol
- Commensal and Probiotics-Host Interactions Laboratory, UMR 1319 Micalis, INRA, 78352, Jouy-en-Josas, France.,UMR 1319 Micalis, AgroparisTech, 78352, Jouy-en-Josas, France.,Department of Gastroenterology and Nutrition, Hôpital SaintAntoine and UPMC University of Paris, 75012, Paris, France.,Equipe AVENIR Gut Microbiota and Immunity, INSERM U1057/UMR CNRS 7203, Université Pierre et Marie Curie 6, 75005, Paris, France
| | - Sylvia H Duncan
- Microbial Ecology Group, Rowett Institute of Nutrition and Health, University of Aberdeen, AB21 9SB, Aberdeen, United Kingdom
| | - Harry J Flint
- Microbial Ecology Group, Rowett Institute of Nutrition and Health, University of Aberdeen, AB21 9SB, Aberdeen, United Kingdom
| | - Hermie J M Harmsen
- Department of Medical Microbiology, University Medical Center Groningen, 9700 RB, Groningen, The Netherlands
| | - Philippe Langella
- Commensal and Probiotics-Host Interactions Laboratory, UMR 1319 Micalis, INRA, 78352, Jouy-en-Josas, France.,UMR 1319 Micalis, AgroparisTech, 78352, Jouy-en-Josas, France
| | - Janneke N Samsom
- Department of Pediatrics, Erasmus Medical Center - Sophia Children's Hospital, 3015 CE, Rotterdam, The Netherlands
| | - Jerry M Wells
- Host-Microbe Interactomics Group, University of Wageningen, 6708 WD, The Netherlands
| |
Collapse
|
147
|
Bellaguarda E, Chang EB. IBD and the gut microbiota--from bench to personalized medicine. Curr Gastroenterol Rep 2015; 17:15. [PMID: 25762474 DOI: 10.1007/s11894-015-0439-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Inflammatory bowel diseases (IBD) are chronic relapsing inflammatory disorders involving the gastrointestinal (GI) tract, which arise from the confluence of genetic, immunological, microbial, and environmental factors. Clinical, genetic, and experimental data support the role of gut microbiota in contributing to the etiopathogenesis of these diseases. In IBD, the development of gut dysbiosis and imbalances in host-microbe relationships contribute to the extent, severity, and chronicity of intestinal inflammation. With continued advances in knowledge, technology, bioinformatics tools, and capabilities to define disease subsets, we will be able to lower risk and improve clinical outcomes in IBD through individualized interventions that restore host-microbial balance. This article provides a critical review of the field, based on the latest clinical and experimental information.
Collapse
Affiliation(s)
- Emanuelle Bellaguarda
- Division of Gastroenterology and Hepatology, Northwestern University, 676 North Saint Clair suite 1400, Chicago, 60611, IL, USA
| | | |
Collapse
|
148
|
Oberc A, Coombes BK. Convergence of External Crohn's Disease Risk Factors on Intestinal Bacteria. Front Immunol 2015; 6:558. [PMID: 26579131 PMCID: PMC4630591 DOI: 10.3389/fimmu.2015.00558] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 10/19/2015] [Indexed: 01/06/2023] Open
Abstract
Crohn’s disease (CD) is an immune-mediated intestinal illness that significantly compromises health in many developed countries. Although definitive causes remain elusive, the required contribution of microbes in the progression of disease has become an accepted concept. Known CD risk factors, such as antibiotic use and acute infectious gastroenteritis, may impact the gut. This concept is now being explored with a view toward understanding the beneficial and unfavorable microbes that may be altered in numbers during such external insults. A comprehensive understanding of the microbial component to CD could be useful clinically as future therapies may focus on preventing risk exposures on susceptible individuals, eliminating harmful microbes, or restoring a protective gut microbiome. Here, we examine how acute infectious gastroenteritis and antibiotic exposure may impact the gut microbiota in the context of inflammation in CD.
Collapse
Affiliation(s)
- Alexander Oberc
- Department of Biochemistry and Biomedical Sciences, McMaster University , Hamilton, ON , Canada ; Michael G. DeGroote Institute for Infectious Disease Research , Hamilton, ON , Canada
| | - Brian K Coombes
- Department of Biochemistry and Biomedical Sciences, McMaster University , Hamilton, ON , Canada ; Michael G. DeGroote Institute for Infectious Disease Research , Hamilton, ON , Canada ; Farncombe Family Digestive Health Research Institute , Hamilton, ON , Canada
| |
Collapse
|
149
|
Kaunitz J, Nayyar P. Bugs, genes, fatty acids, and serotonin: Unraveling inflammatory bowel disease? F1000Res 2015; 4:F1000 Faculty Rev-1146. [PMID: 27508055 PMCID: PMC4963018 DOI: 10.12688/f1000research.6456.1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/30/2015] [Indexed: 12/13/2022] Open
Abstract
The annual incidence of the inflammatory bowel diseases (IBDs) ulcerative colitis and Crohn's disease has increased at an alarming rate. Although the specific pathophysiology underlying IBD continues to be elusive, it is hypothesized that IBD results from an aberrant and persistent immune response directed against microbes or their products in the gut, facilitated by the genetic susceptibility of the host and intrinsic alterations in mucosal barrier function. In this review, we will describe advances in the understanding of how the interaction of host genetics and the intestinal microbiome contribute to the pathogenesis of IBD, with a focus on bacterial metabolites such as short chain fatty acids (SCFAs) as possible key signaling molecules. In particular, we will describe alterations of the intestinal microbiota in IBD, focusing on how genetic loci affect the gut microbial phylogenetic distribution and the production of their major microbial metabolic product, SCFAs. We then describe how enteroendocrine cells and myenteric nerves express SCFA receptors that integrate networks such as the cholinergic and serotonergic neural systems and the glucagon-like peptide hormonal pathway, to modulate gut inflammation, permeability, and growth as part of an integrated model of IBD pathogenesis. Through this integrative approach, we hope that novel hypotheses will emerge that will be tested in reductionist, hypothesis-driven studies in order to examine the interrelationship of these systems in the hope of better understanding IBD pathogenesis and to inform novel therapies.
Collapse
Affiliation(s)
- Jonathan Kaunitz
- Medical Service, West Los Angeles VAMC, Los Angeles, CA, 90073, USA
- Department of Medicine, David Geffen School of Medicine, Los Angeles, CA, 90095, USA
- Department of Surgery, David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Piyush Nayyar
- Medical Service, West Los Angeles VAMC, Los Angeles, CA, 90073, USA
- Department of Medicine, David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| |
Collapse
|
150
|
Torres-Maravilla E, Lenoir M, Mayorga-Reyes L, Allain T, Sokol H, Langella P, Sánchez-Pardo ME, Bermúdez-Humarán LG. Identification of novel anti-inflammatory probiotic strains isolated from pulque. Appl Microbiol Biotechnol 2015; 100:385-396. [DOI: 10.1007/s00253-015-7049-4] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 09/22/2015] [Accepted: 09/29/2015] [Indexed: 02/06/2023]
|