401
|
Romarheim OH, Hetland DL, Skrede A, Øverland M, Mydland LT, Landsverk T. Prevention of soya-induced enteritis in Atlantic salmon (Salmo salar) by bacteria grown on natural gas is dose dependent and related to epithelial MHC II reactivity and CD8α+ intraepithelial lymphocytes. Br J Nutr 2013; 109:1062-70. [PMID: 22813713 DOI: 10.1017/s0007114512002899] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
An experiment was carried out to study the preventive effect of bacterial meal (BM) produced from natural gas against plant-induced enteropathy in Atlantic salmon (Salmo salar). Salmon were fed a diet based on fish meal (FM) or seven diets with 200 g/kg solvent-extracted soyabean meal (SBM) to induce enteritis in combination with increasing levels of BM from 0 to 300 g/kg. Salmon fed a SBM-containing diet without BM developed typical SBM-induced enteritis. The enteritis gradually disappeared with increasing inclusion of BM. By morphometry, no significant (P>0.05) differences in the size of stretches stained for proliferating cell nuclear antigen were found with 150 g/kg BM compared with the FM diet. Increasing BM inclusion caused a gradual decline in the number of cluster of differentiation 8 α positive (CD8α+) intraepithelial lymphocytes, and fish fed BM at 200 g/kg or higher revealed no significant difference from the FM diet. Histological sections stained with antibody for MHC class II (MHC II) showed that fish with intestinal inflammation had more MHC II-reactive cells in the lamina propria and submucosa, but less in the epithelium and brush border, compared with fish without inflammation. There were no significant (P>0.05) differences in growth among the diets, but the highest levels of BM slightly reduced protein digestibility and increased the weight of the distal intestine. In conclusion, the prevention of SBM-induced enteritis by BM is dose dependent and related to intestinal levels of MHC II- and CD8α-reactive cells.
Collapse
Affiliation(s)
- Odd H Romarheim
- Department of Animal and Aquacultural Sciences, Aquaculture Protein Centre, CoE, Norwegian University of Life Sciences, Ås, Norway.
| | | | | | | | | | | |
Collapse
|
402
|
Tomosada Y, Villena J, Murata K, Chiba E, Shimazu T, Aso H, Iwabuchi N, Xiao JZ, Saito T, Kitazawa H. Immunoregulatory effect of bifidobacteria strains in porcine intestinal epithelial cells through modulation of ubiquitin-editing enzyme A20 expression. PLoS One 2013; 8:e59259. [PMID: 23555642 PMCID: PMC3608626 DOI: 10.1371/journal.pone.0059259] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Accepted: 02/13/2013] [Indexed: 12/20/2022] Open
Abstract
Background We previously showed that evaluation of anti-inflammatory activities of lactic acid bacteria in porcine intestinal epithelial (PIE) cells is useful for selecting potentially immunobiotic strains. Objective The aims of the present study were: i) to select potentially immunomodulatory bifidobacteria that beneficially modulate the Toll-like receptor (TLR)-4-triggered inflammatory response in PIE cells and; ii) to gain insight into the molecular mechanisms involved in the anti-inflammatory effect of immunobiotics by evaluating the role of TLR2 and TLR negative regulators in the modulation of proinflammatory cytokine production and activation of mitogen-activated protein kinase (MAPK) and nuclear factor-κB (NF-κB) pathways in PIE cells. Results Bifidobacteria longum BB536 and B. breve M-16V strains significantly downregulated levels of interleukin (IL)-8, monocyte chemotactic protein (MCP)-1 and IL-6 in PIE cells challenged with heat-killed enterotoxigenic Escherichia coli. Moreover, BB536 and M-16V strains attenuated the proinflammatory response by modulating the NF-κB and MAPK pathways. In addition, our findings provide evidence for a key role for the ubiquitin-editing enzyme A20 in the anti-inflammatory effect of immunobiotic bifidobacteria in PIE cells. Conclusions We show new data regarding the mechanism involved in the anti-inflammatory effect of immunobiotics. Several strains with immunoregulatory capabilities used a common mechanism to induce tolerance in PIE cells. Immunoregulatory strains interacted with TLR2, upregulated the expression of A20 in PIE cells, and beneficially modulated the subsequent TLR4 activation by reducing the activation of MAPK and NF-κB pathways and the production of proinflammatory cytokines. We also show that the combination of TLR2 activation and A20 induction can be used as biomarkers to screen and select potential immunoregulatory bifidobacteria strains.
Collapse
Affiliation(s)
- Yohsuke Tomosada
- Food and Feed Immunology Group, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Julio Villena
- Food and Feed Immunology Group, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
- Laboratory of Clinical and Experimental Biochemistry, Reference Centre for Lactobacilli (CERELA-CONICET), Tucuman, Argentina
| | - Kozue Murata
- Food and Feed Immunology Group, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Eriko Chiba
- Food and Feed Immunology Group, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Tomoyuki Shimazu
- Laboratory of Animal Breeding and Genetics, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Hisashi Aso
- Cell Biology Laboratory, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Noriyuki Iwabuchi
- Food Science and Technology Institute, Morinaga Milk Industry Co. Ltd, Zama, Kanagawa, Japan
| | - Jin-zhong Xiao
- Food Science and Technology Institute, Morinaga Milk Industry Co. Ltd, Zama, Kanagawa, Japan
| | - Tadao Saito
- Food and Feed Immunology Group, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Haruki Kitazawa
- Food and Feed Immunology Group, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
- * E-mail:
| |
Collapse
|
403
|
Abstract
Commensal bacteria are necessary for the development and maintenance of a healthy immune system. Harnessing the ability of microbiota to affect host immunity is considered an important therapeutic strategy for many mucosal and nonmucosal immune-related conditions, such as inflammatory bowel diseases (IBDs), celiac disease, metabolic syndrome, diabetes, and microbial infections. In addition to well-established immunostimulatory effects of the microbiota, the presence of individual mutualistic commensal bacteria with immunomodulatory effects has been described. These organisms are permanent members of the commensal microbiota and affect host immune homeostasis in specific ways. Identification of individual examples of such immunomodulatory commensals and understanding their mechanisms of interaction with the host will be invaluable in designing therapeutic strategies to reverse intestinal dysbiosis and recover immunological homeostasis.
Collapse
Affiliation(s)
- Ivaylo I Ivanov
- Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY 10032, USA.
| | | |
Collapse
|
404
|
Caricilli AM, Saad MJA. The role of gut microbiota on insulin resistance. Nutrients 2013; 5:829-51. [PMID: 23482058 PMCID: PMC3705322 DOI: 10.3390/nu5030829] [Citation(s) in RCA: 150] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Revised: 01/10/2013] [Accepted: 01/15/2013] [Indexed: 12/12/2022] Open
Abstract
The development of obesity and insulin resistance has been extensively studied in the last decades, but the mechanisms underlying these alterations are still not completely understood. The gut microbiota has been identified as a potential contributor to metabolic diseases. It has been shown that obese individuals present different proportions of bacterial phyla compared with lean individuals, with an increase in Firmicutes and Actinobacteria and a decrease in Bacteroidetes. This alteration seems to interfere with intestinal permeability, increasing the absorption of lipopolysaccharide (LPS), which reaches circulation and initiates activation of Toll-like receptor (TLR) 4 and 2 and LPS receptor CD14, leading to increased activation of inflammatory pathways. With these activations, an impairment of the insulin signaling is observed, with decreased phosphorylation of the insulin receptor, insulin receptor substrate (IRS) and Akt, as well as increased inhibitory serine phosphorylation of IRS-1. Altered proportions of bacterial phyla have also been demonstrated to interfere with host’s biochemical pathways, increasing energy extraction and depot in adipose tissue. Therefore, understanding the mechanisms by which the alteration in the gut microbiota produces different signaling activations and phenotype changes may offer an interesting opportunity for the treatment of obesity and type 2 diabetes.
Collapse
Affiliation(s)
- Andrea M. Caricilli
- Department of Immunology, ICB IV, University of São Paulo, Av. Prof. Lineu Prestes, 2415, Cidade Universitária, São Paulo, SP, Brazil; E-Mail:
| | - Mario J. A. Saad
- Department of Internal Medicine, State University of Campinas, Rua Tessália Vieira de Camargo, 126, Cidade Universitária, Campinas, SP, Brazil
- Author to whom correspondence should be addressed; E-Mail: ; Tel./Fax: +55-19-35218950
| |
Collapse
|
405
|
Nelson RK, Poroyko V, Morowitz MJ, Liu D, Alverdy JC. Effect of dietary monosaccharides on Pseudomonas aeruginosa virulence. Surg Infect (Larchmt) 2013; 14:35-42. [PMID: 23451729 DOI: 10.1089/sur.2011.063] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Pseudomonas aeruginosa is an opportunistic, gram-negative pathogen associated with many hospital-acquired infections and disease states. In particular, P. aeruginosa has been identified as a crucial factor in the pathogenesis of neonatal necrotizing enterocolitis (NEC). This condition presents more frequently in infants fed a formula-based diet, which may be a result of the specific monosaccharide content of this diet. We hypothesized that P. aeruginosa would express virulence genes differentially when exposed to monosaccharides present in formula versus those in human milk. METHODS Using the results of a metabolomics study on infant diets and their resulting fecal samples, we identified several monosaccharides that distinguished milk from formula diets. Of these compounds, four were found to be metabolized by P. aeruginosa. We subsequently grew P. aeruginosa in tryptic soy broth (TSB) supplemented with these four monosaccharides and used quantitative reverse transcriptase-polymerase chain reaction to measure the expression of 59 major P. aeruginosa virulence genes. The results were standardized to an external control of P. aeruginosa grown in TSB alone. RESULTS P. aeruginosa did not respond differentially to the monosaccharides after 6 h of growth. However, after 24 h, the organism grown in arabinose (present in formula), xylose (present in human milk), and galactose (present in both formula and feces from milk-fed infants) displayed a significant increase in the expression of virulence genes in all categories. In contrast, P. aeruginosa grown in mannose (present in the feces of milk-fed infants) displayed a significant decrease in virulence gene expression. CONCLUSION These results demonstrate the importance of nutrient content on the relative expression of virulence genes in pathogens that colonize commonly the gut of infants. Understanding the effect of current dietary formulas on virulence gene expression in various gut-colonizing pathogens may present a new approach to elucidating the differences between human milk and formula in the development of NEC.
Collapse
Affiliation(s)
- Ryan K Nelson
- Department of Surgery, University of Chicago Pritzker School of Medicine, Chicago, Illinois 60637, USA
| | | | | | | | | |
Collapse
|
406
|
Maccaferri S, Candela M, Turroni S, Centanni M, Severgnini M, Consolandi C, Cavina P, Brigidi P. IBS-associated phylogenetic unbalances of the intestinal microbiota are not reverted by probiotic supplementation. Gut Microbes 2013; 3:406-13. [PMID: 22713265 DOI: 10.4161/gmic.21009] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
IBS is a prevalent functional gastrointestinal disorder, in which the microbiota has been demonstrated to play a role. An increasing number of studies have suggested how probiotics may alleviate IBS symptoms and several mechanisms of action have been proposed. In the present study we characterized the intestinal microbiota of 19 subjects suffering from diagnosed IBS using a fully validated High Taxonomic Fingerprint Microbiota Array (HTF-Microbi.Array). We demonstrated that the IBS microbiota is different from that of healthy individuals due to an unbalance in a number of commensal species, with an increase in relative abundance of lactobacilli, B. cereus and B. clausii, bifidobacteria, Clostridium cluster IX and E. rectale, and a decrease in abundance of Bacteroides/Prevotella group and Veillonella genus. Additionally, we demonstrated that some bacterial groups of the human intestinal microbiota, recently defined as pathobionts, are increased in concentration in the IBS microbiota. Furthermore, we aimed at investigating if the daily administration of a novel probiotic yogurt containing B. animalis subsp lactis Bb12 and K. marxianus B0399, recently demonstrated to have beneficial effects in the management of IBS symptoms, could impact on the biostructure of IBS microbiota, modulating its composition to counteract putative dysbiosis found in IBS subjects. Notably, we demonstrated that the beneficial effects associated to the probiotic preparation are not related to significant modifications in the composition of the human intestinal microbiota.
Collapse
Affiliation(s)
- Simone Maccaferri
- Department of Pharmaceutical Sciences, University of Bologna, Bologna, Italy
| | | | | | | | | | | | | | | |
Collapse
|
407
|
|
408
|
Ellis RJ, Bruce KD, Jenkins C, Stothard JR, Ajarova L, Mugisha L, Viney ME. Comparison of the distal gut microbiota from people and animals in Africa. PLoS One 2013; 8:e54783. [PMID: 23355898 PMCID: PMC3552852 DOI: 10.1371/journal.pone.0054783] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2011] [Accepted: 12/17/2012] [Indexed: 01/20/2023] Open
Abstract
The gut microbiota plays a key role in the maintenance of healthy gut function as well as many other aspects of health. High-throughput sequence analyses have revealed the composition of the gut microbiota, showing that there is a core signature to the human gut microbiota, as well as variation in its composition between people. The gut microbiota of animals is also being investigated. We are interested in the relationship between bacterial taxa of the human gut microbiota and those in the gut microbiota of domestic and semi-wild animals. While it is clear that some human gut bacterial pathogens come from animals (showing that human--animal transmission occurs), the extent to which the usually non-pathogenic commensal taxa are shared between humans and animals has not been explored. To investigate this we compared the distal gut microbiota of humans, cattle and semi-captive chimpanzees in communities that are geographically sympatric in Uganda. The gut microbiotas of these three host species could be distinguished by the different proportions of bacterial taxa present. We defined multiple operational taxonomic units (OTUs) by sequence similarity and found evidence that some OTUs were common between human, cattle and chimpanzees, with the largest number of shared OTUs occurring between chimpanzees and humans, as might be expected with their close physiological similarity. These results show the potential for the sharing of usually commensal bacterial taxa between humans and other animals. This suggests that further investigation of this phenomenon is needed to fully understand how it drives the composition of human and animal gut microbiotas.
Collapse
Affiliation(s)
- Richard J Ellis
- Specialist Scientific Support Department, Animal Health and Veterinary Laboratories Agency, Addlestone, Surrey, United Kingdom.
| | | | | | | | | | | | | |
Collapse
|
409
|
Amelioration of experimental autoimmune encephalomyelitis by probiotic mixture is mediated by a shift in T helper cell immune response. Clin Immunol 2013; 146:217-27. [PMID: 23416238 DOI: 10.1016/j.clim.2013.01.001] [Citation(s) in RCA: 146] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Revised: 12/24/2012] [Accepted: 01/08/2013] [Indexed: 12/13/2022]
Abstract
The immunomodulatory effect of probiotics has been shown mainly in gastro-intestinal immune disorders and little information is available on the inflammation of central nervous system. Recently we reported that IRT5 probiotics, a mixture of 5 probiotics, could suppress diverse experimental inflammatory disorders. In this study, we evaluated the prophylactic and therapeutic effects of IRT5 probiotics in experimental autoimmune encephalomyelitis (EAE), a T cell mediated inflammatory autoimmune disease of the central nervous system. Pretreatment of IRT5 probiotics before disease induction significantly suppressed EAE development. In addition, treatment with IRT5 probiotics to the ongoing EAE delayed the disease onset. Administration of IRT5 probiotics inhibited the pro-inflammatory Th1/Th17 polarization, while inducing IL10(+) producing or/and Foxp3(+) regulatory T cells, both in the peripheral immune system and at the site of inflammation. Collectively, our data suggest that IRT5 probiotics could be applicable to modulate T cell mediated neuronal autoimmune diseases, including multiple sclerosis.
Collapse
|
410
|
Schachtschneider KM, Yeoman CJ, Isaacson RE, White BA, Schook LB, Pieters M. Modulation of systemic immune responses through commensal gastrointestinal microbiota. PLoS One 2013; 8:e53969. [PMID: 23326551 PMCID: PMC3543314 DOI: 10.1371/journal.pone.0053969] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Accepted: 12/05/2012] [Indexed: 12/29/2022] Open
Abstract
Colonization of the gastrointestinal (GI) tract is initiated during birth and continually seeded from the individual's environment. Gastrointestinal microorganisms play a central role in developing and modulating host immune responses and have been the subject of investigation over the last decades. Animal studies have demonstrated the impact of GI tract microbiota on local gastrointestinal immune responses; however, the full spectrum of action of early gastrointestinal tract stimulation and subsequent modulation of systemic immune responses is poorly understood. This study explored the utility of an oral microbial inoculum as a therapeutic tool to affect porcine systemic immune responses. For this study a litter of 12 pigs was split into two groups. One group of pigs was inoculated with a non-pathogenic oral inoculum (modulated), while another group (control) was not. DNA extracted from nasal swabs and fecal samples collected throughout the study was sequenced to determine the effects of the oral inoculation on GI and respiratory microbial communities. The effects of GI microbial modulation on systemic immune responses were evaluated by experimentally infecting with the pathogen Mycoplasma hyopneumoniae. Coughing levels, pathology, toll-like receptors 2 and 6, and cytokine production were measured throughout the study. Sequencing results show a successful modulation of the GI and respiratory microbiomes through oral inoculation. Delayed type hypersensitivity responses were stronger (p = 0.07), and the average coughing levels and respiratory TNF-α variance were significantly lower in the modulated group (p<0.0001 and p = 0.0153, respectively). The M. hyopneumoniae infection study showed beneficial effects of the oral inoculum on systemic immune responses including antibody production, severity of infection and cytokine levels. These results suggest that an oral microbial inoculation can be used to modulate microbial communities, as well as have a beneficial effect on systemic immune responses as demonstrated with M. hyopneumoniae infection.
Collapse
Affiliation(s)
- Kyle M. Schachtschneider
- Department of Animal Sciences, University of Illinois, Urbana, Illinois, United States of America
| | - Carl J. Yeoman
- Institute for Genomic Biology, University of Illinois, Urbana, Illinois, United States of America
- Department of Animal and Range Sciences, Montana State University, Montana, United States of America
| | - Richard E. Isaacson
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, Minnesota, United States of America
| | - Bryan A. White
- Department of Animal Sciences, University of Illinois, Urbana, Illinois, United States of America
- Institute for Genomic Biology, University of Illinois, Urbana, Illinois, United States of America
| | - Lawrence B. Schook
- Department of Animal Sciences, University of Illinois, Urbana, Illinois, United States of America
- Institute for Genomic Biology, University of Illinois, Urbana, Illinois, United States of America
| | - Maria Pieters
- Institute for Genomic Biology, University of Illinois, Urbana, Illinois, United States of America
| |
Collapse
|
411
|
Hara N, Alkanani AK, Ir D, Robertson CE, Wagner BD, Frank DN, Zipris D. The role of the intestinal microbiota in type 1 diabetes. Clin Immunol 2012; 146:112-9. [PMID: 23314185 DOI: 10.1016/j.clim.2012.12.001] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Revised: 11/30/2012] [Accepted: 12/01/2012] [Indexed: 02/07/2023]
Abstract
The digestive tract hosts trillions of bacteria that interact with the immune system and can influence the balance between pro-inflammatory and regulatory immune responses. Recent studies suggest that alterations in the composition of the intestinal microbiota may be linked with the development of type 1 diabetes (T1D). Data from the biobreeding diabetes prone (BBDP) and the LEW1.WR1 models of T1D support the hypothesis that intestinal bacteria may be involved in early disease mechanisms. The data indicate that cross-talk between the gut microbiota and the innate immune system may be involved in islet destruction. Whether a causal link between intestinal microbiota and T1D exists, the identity of the bacteria and the mechanism whereby they promote the disease remain to be examined. A better understanding of the interplay between microbes and innate immune pathways in early disease stages holds promise for the design of immune interventions and disease prevention in genetically susceptible individuals.
Collapse
Affiliation(s)
- Naoko Hara
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, CO 80045, USA
| | | | | | | | | | | | | |
Collapse
|
412
|
Le Roux D, Niedergang F. New insights into antigen encounter by B cells. Immunobiology 2012; 217:1285-91. [DOI: 10.1016/j.imbio.2012.07.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Revised: 07/10/2012] [Accepted: 07/14/2012] [Indexed: 01/01/2023]
|
413
|
Atkinson MA, Chervonsky A. Does the gut microbiota have a role in type 1 diabetes? Early evidence from humans and animal models of the disease. Diabetologia 2012; 55:2868-77. [PMID: 22875196 PMCID: PMC3496388 DOI: 10.1007/s00125-012-2672-4] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2012] [Accepted: 06/26/2012] [Indexed: 02/07/2023]
Abstract
Despite years of appreciating the potential role of environment to influence the pathogenesis of type 1 diabetes, specific agents or mechanisms serving in such a capacity remain ill defined. This is exceedingly disappointing as the identification of factors capable of modulating the disease, either as triggers or regulators of the autoimmune response underlying type 1 diabetes, would not only provide clues as to why the disorder develops but, in addition, afford opportunities for improved biomarkers of disease activity and the potential to design novel therapeutics capable of disease abatement. Recent improvements in sequencing technologies, combined with increasing appreciation of the role of innate and mucosal immunity in human disease, have stirred strong interest in what is commonly referred to as the 'gut microbiota'. The gut (or intestinal) microbiota is an exceedingly complex microenvironment that is intimately linked with the immune system, including the regulation of immune responses. After evaluating evidence supporting a role for environment in type 1 diabetes, this review will convey current notions for contributions of the gut microbiota to human health and disease, including information gleaned from studies of humans and animal models for this autoimmune disorder.
Collapse
Affiliation(s)
- M A Atkinson
- Department of Pathology, University of Florida, College of Medicine, 1600 SW Archer Road, Gainesville, FL 32610-0275, USA.
| | | |
Collapse
|
414
|
Olier M, Marcq I, Salvador-Cartier C, Secher T, Dobrindt U, Boury M, Bacquié V, Penary M, Gaultier E, Nougayrède JP, Fioramonti J, Oswald E. Genotoxicity of Escherichia coli Nissle 1917 strain cannot be dissociated from its probiotic activity. Gut Microbes 2012; 3:501-9. [PMID: 22895085 PMCID: PMC3495787 DOI: 10.4161/gmic.21737] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Oral administration of the probiotic bacterium Escherichia coli Nissle 1917 improves chronic inflammatory bowel diseases, but the molecular basis for this therapeutic efficacy is unknown. E. coli Nissle 1917 harbors a cluster of genes coding for the biosynthesis of hybrid nonribosomal peptide-polyketide(s). This biosynthetic pathway confers the ability for bacteria to induce DNA double strand breaks in eukaryotic cells. Here we reveal that inactivation of the clbA gene within this genomic island abrogated the ability for the strain to induce DNA damage and chromosomal abnormalities in non-transformed cultured rat intestinal epithelial cells but is required for the probiotic activity of E. coli Nissle 1917. Thus, evaluation of colitis severity induced in rodent fed with E. coli Nissle 1917 or an isogenic non-genotoxic mutant demonstrated the need for a functional biosynthetic pathway both in the amelioration of the disease and in the modulation of cytokine expression. Feeding rodents with a complemented strain for which genotoxicity was restored confirmed that this biosynthetic pathway contributes to the health benefits of the probiotic by modulating its immunomodulatory properties. Our data provide additional evidence for the benefit of this currently used probiotic in colitis but remind us that an efficient probiotic may also have side effects as any other medication.
Collapse
Affiliation(s)
- Maïwenn Olier
- Neuro-gastroenterologie et Nutrition; UMR INRA/ENVT 1331; Toulouse, France,INRA; USC 1360; Toulouse, France,Inserm; UMR1043; Toulouse, France,CNRS; UMR5282; Toulouse, France,Université de Toulouse; UPS, Centre de Physiopathologie de Toulouse Purpan (CPTP); Toulouse, France,Correspondence to: Maïwenn Olier, and Eric Oswald,
| | - Ingrid Marcq
- INRA; USC 1360; Toulouse, France,Inserm; UMR1043; Toulouse, France,CNRS; UMR5282; Toulouse, France,Université de Toulouse; UPS, Centre de Physiopathologie de Toulouse Purpan (CPTP); Toulouse, France
| | | | - Thomas Secher
- INRA; USC 1360; Toulouse, France,Inserm; UMR1043; Toulouse, France,CNRS; UMR5282; Toulouse, France,Université de Toulouse; UPS, Centre de Physiopathologie de Toulouse Purpan (CPTP); Toulouse, France
| | | | - Michèle Boury
- INRA; USC 1360; Toulouse, France,Inserm; UMR1043; Toulouse, France,CNRS; UMR5282; Toulouse, France,Université de Toulouse; UPS, Centre de Physiopathologie de Toulouse Purpan (CPTP); Toulouse, France
| | - Valérie Bacquié
- Neuro-gastroenterologie et Nutrition; UMR INRA/ENVT 1331; Toulouse, France
| | - Marie Penary
- INRA; USC 1360; Toulouse, France,Inserm; UMR1043; Toulouse, France,CNRS; UMR5282; Toulouse, France,Université de Toulouse; UPS, Centre de Physiopathologie de Toulouse Purpan (CPTP); Toulouse, France
| | - Eric Gaultier
- Neuro-gastroenterologie et Nutrition; UMR INRA/ENVT 1331; Toulouse, France
| | - Jean-Philippe Nougayrède
- INRA; USC 1360; Toulouse, France,Inserm; UMR1043; Toulouse, France,CNRS; UMR5282; Toulouse, France,Université de Toulouse; UPS, Centre de Physiopathologie de Toulouse Purpan (CPTP); Toulouse, France
| | - Jean Fioramonti
- Neuro-gastroenterologie et Nutrition; UMR INRA/ENVT 1331; Toulouse, France
| | - Eric Oswald
- INRA; USC 1360; Toulouse, France,Inserm; UMR1043; Toulouse, France,CNRS; UMR5282; Toulouse, France,Université de Toulouse; UPS, Centre de Physiopathologie de Toulouse Purpan (CPTP); Toulouse, France,CHU Toulouse; Hôpital Purpan; Service de bactériologie-Hygiène; Toulouse, France,Correspondence to: Maïwenn Olier, and Eric Oswald,
| |
Collapse
|
415
|
Effects of crude oil, dispersant, and oil-dispersant mixtures on human fecal microbiota in an in vitro culture system. mBio 2012; 3:mBio.00376-12. [PMID: 23093387 PMCID: PMC3482501 DOI: 10.1128/mbio.00376-12] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The Deepwater Horizon oil spill of 2010 raised concerns that dispersant and dispersed oil, as well as crude oil itself, could contaminate shellfish and seafood habitats with hazardous residues that had potential implications for human health and the ecosystem. However, little is known about the effects of crude oil and dispersant on the human fecal microbiota. The aim of this research was to evaluate the potential effects of Deepwater Horizon crude oil, Corexit 9500 dispersant, and their combination on human fecal microbial communities, using an in vitro culture test system. Fecal specimens from healthy adult volunteers were made into suspensions, which were then treated with oil, dispersant, or oil-dispersant mixtures under anaerobic conditions in an in vitro culture test system. Perturbations of the microbial community, compared to untreated control cultures, were assessed using denaturing gradient gel electrophoresis (DGGE), real-time PCR, and pyrosequencing methods. DGGE and pyrosequencing analysis showed that oil-dispersant mixtures reduced the diversity of fecal microbiota from all individuals. Real-time PCR results indicated that the copy numbers of 16S rRNA genes in cultures treated with dispersed oil or oil alone were significantly lower than those in control incubations. The abundance of the Bacteroidetes decreased in crude oil-treated and dispersed-oil-treated cultures, while the Proteobacteria increased in cultures treated with dispersed oil. In conclusion, the human fecal microbiota was affected differently by oil and dispersed oil, and the influence of dispersed oil was significantly greater than that of either oil or dispersant alone compared to control cultures. There have been concerns whether human health is adversely affected by exposure to spilled crude oil, which contains regulated carcinogens, such as polycyclic aromatic hydrocarbons. In this study, we determined the effect of BP Deepwater Horizon crude oil and oil dispersant on the human intestinal microbiota, since there is the potential that low-level residues of petrochemicals could contaminate seafood. The results of this study will increase our understanding of the ecophysiological changes in the microbial communities of the human gastrointestinal tract with respect to crude oil exposure.
Collapse
|
416
|
Natividad JMM, Verdu EF. Modulation of intestinal barrier by intestinal microbiota: pathological and therapeutic implications. Pharmacol Res 2012; 69:42-51. [PMID: 23089410 DOI: 10.1016/j.phrs.2012.10.007] [Citation(s) in RCA: 296] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Revised: 10/11/2012] [Accepted: 10/13/2012] [Indexed: 02/07/2023]
Abstract
Mammals and their intestinal microbiota peacefully coexist in a mutualistic relationship. Commensal bacteria play an active role in shaping and modulating physiological processes in the host, which include, but are not restricted to, the immune system and the intestinal barrier. Both play a crucial role in containing intestinal bacteria and other potentially noxious luminal antigens within the lumen and mucosal compartment. Although mutualism defines the relationship between the host and the intestinal microbiota, disruptions in this equilibrium may promote disease. Thus, alterations in gut microbiota (dysbiosis) have been linked to the recent increased expression of obesity, allergy, autoimmunity, functional and inflammatory disorders such as irritable bowel syndrome (IBS) and inflammatory bowel disease (IBD). In this article, we review the evidence supporting a role of gut microbiota in regulating intestinal barrier function. We discuss the hypothesis that microbial factors can modulate the barrier in ways that can prevent or promote gastrointestinal disease. A better understanding of the role of the intestinal microbiota in maintaining a functional intestinal barrier may help develop targeted strategies to prevent and treat disease.
Collapse
Affiliation(s)
- Jane M M Natividad
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Canada
| | | |
Collapse
|
417
|
Rol N, Favre L, Benyacoub J, Corthésy B. The role of secretory immunoglobulin A in the natural sensing of commensal bacteria by mouse Peyer's patch dendritic cells. J Biol Chem 2012; 287:40074-82. [PMID: 23027876 DOI: 10.1074/jbc.m112.405001] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The mammalian gastrointestinal (GI) tract harbors a diverse population of commensal species collectively known as the microbiota, which interact continuously with the host. From very early in life, secretory IgA (SIgA) is found in association with intestinal bacteria. It is considered that this helps to ensure self-limiting growth of the microbiota and hence participates in symbiosis. However, the importance of this association in contributing to the mechanisms ensuring natural host-microorganism communication is in need of further investigation. In the present work, we examined the possible role of SIgA in the transport of commensal bacteria across the GI epithelium. Using an intestinal loop mouse model and fluorescently labeled bacteria, we found that entry of commensal bacteria in Peyer's patches (PP) via the M cell pathway was mediated by their association with SIgA. Preassociation of bacteria with nonspecific SIgA increased their dynamics of entry and restored the reduced transport observed in germ-free mice known to have a marked reduction in intestinal SIgA production. Selective SIgA-mediated targeting of bacteria is restricted to the tolerogenic CD11c(+)CD11b(+)CD8(-) dendritic cell subset located in the subepithelial dome region of PPs, confirming that the host is not ignorant of its resident commensals. In conclusion, our work supports the concept that SIgA-mediated monitoring of commensal bacteria targeting dendritic cells in the subepithelial dome region of PPs represents a mechanism whereby the host mucosal immune system controls the continuous dialogue between the host and commensal bacteria.
Collapse
Affiliation(s)
- Nicolas Rol
- Research and Development Laboratory, Division of Immunology and Allergy, Centre Hospitalier Universitaire Vaudois, 1011 Lausanne, Switzerland
| | | | | | | |
Collapse
|
418
|
Lang ML, Zhu L, Kreth J. Keeping the bad bacteria in check: interactions of the host immune system with oral cavity biofilms. ACTA ACUST UNITED AC 2012. [DOI: 10.1111/j.1601-1546.2012.00278.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
419
|
Veldhoen M, Brucklacher-Waldert V. Dietary influences on intestinal immunity. Nat Rev Immunol 2012; 12:696-708. [DOI: 10.1038/nri3299] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
420
|
Hara N, Alkanani AK, Ir D, Robertson CE, Wagner BD, Frank DN, Zipris D. Prevention of virus-induced type 1 diabetes with antibiotic therapy. THE JOURNAL OF IMMUNOLOGY 2012; 189:3805-14. [PMID: 22988033 DOI: 10.4049/jimmunol.1201257] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Microbes were hypothesized to play a key role in the progression of type 1 diabetes (T1D). We used the LEW1.WR1 rat model of Kilham rat virus (KRV)-induced T1D to test the hypothesis that the intestinal microbiota is involved in the mechanism leading to islet destruction. Treating LEW1.WR1 rats with KRV and a combination of trimethoprim and sulfamethoxazole (Sulfatrim) beginning on the day of infection protected the rats from insulitis and T1D. Pyrosequencing of bacterial 16S rRNA and quantitative RT-PCR indicated that KRV infection resulted in a transient increase in the abundance of Bifidobacterium spp. and Clostridium spp. in fecal samples from day 5- but not day 12-infected versus uninfected animals. Similar alterations in the gut microbiome were observed in the jejunum of infected animals on day 5. Treatment with Sulfatrim restored the level of intestinal Bifidobacterium spp. and Clostridium spp. We also observed that virus infection induced the expression of KRV transcripts and the rapid upregulation of innate immune responses in Peyer's patches and pancreatic lymph nodes. However, antibiotic therapy reduced the virus-induced inflammation as reflected by the presence of lower amounts of proinflammatory molecules in both the Peyer's patches and pancreatic lymph nodes. Finally, Sulfatrim treatment reduced the number of B cells in Peyer's patches and downmodulated adaptive immune responses to KRV, but did not interfere with antiviral Ab responses or viral clearance from the spleen, pancreatic lymph nodes, and serum. The data suggest that gut microbiota may be involved in promoting virus-induced T1D in the LEW1.WR1 rat model.
Collapse
Affiliation(s)
- Naoko Hara
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, CO 80045, USA
| | | | | | | | | | | | | |
Collapse
|
421
|
The immune response to tuberculosis infection in the setting of Helicobacter pylori and helminth infections. Epidemiol Infect 2012; 141:1232-43. [PMID: 22954328 DOI: 10.1017/s0950268812001823] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
We screened 176 healthy, adult (aged 18-55 years) US refugees from tuberculosis (TB)-endemic countries to evaluate whether cytokine responses to latent TB infection (LTBI) are modified in the setting of concurrent H. pylori and helminth infection. As measured by the Quantiferon-TB GOLD interferon-γ release assay, a total 38 (22%) subjects had LTBI, of which 28 (74%) also were H. pylori seropositive and/or helminth infected. Relative to ten subjects with LTBI only, 16 subjects with concurrent H. pylori infection had significantly elevated levels of IFN-γ, and nine subjects with both H. pylori and helminth infection had significantly elevated levels of IFN-γ, IL-2, IL-13, and IL-5. H. pylori is associated with enhanced IFN-γ responses to TB, even in the setting of concurrent helminth infection. Efficacy of TB vaccines may vary with the co-existence of these three infections in the developing world.
Collapse
|
422
|
Daughton CG, Ruhoy IS. Green pharmacy and pharmEcovigilance: prescribing and the planet. Expert Rev Clin Pharmacol 2012; 4:211-32. [PMID: 22115404 DOI: 10.1586/ecp.11.6] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Active pharmaceutical ingredients (APIs) are ubiquitous environmental contaminants, resulting primarily from excretion and bathing and from disposal of leftover drugs by consumers and healthcare facilities. Although prudent disposal of leftover drugs has attracted the most attention for reducing API levels in the aquatic environment, a more effective approach would prevent the generation of leftover drugs in the first place. Many aspects of the practice of medicine and pharmacy can be targeted for reducing environmental contamination by APIs. These same modifications--focused on treating humans and the environment as a single, integral patient--could also have collateral outcomes with improved therapeutic outcomes, and with a reduced incidence of unintended poisonings, drug interactions and drug diversion, and lower consumer costs.
Collapse
Affiliation(s)
- Christian G Daughton
- Office of Research and Development, US Environmental Protection Agency, Las Vegas, NV, USA.
| | | |
Collapse
|
423
|
Size and dynamics of mucosal and peripheral IL-17A+ T-cell pools in pediatric age, and their disturbance in celiac disease. Mucosal Immunol 2012; 5:513-23. [PMID: 22569303 DOI: 10.1038/mi.2012.26] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Mucosal interleukin (IL)-17A-producing T cells contribute to protective antimicrobial responses and to epithelial barrier integrity; their role in celiac disease (CD) is debated. We analyzed the frequency and developmental dynamics of mucosal (intraepithelial lymphocytes (IEL)) and circulating (peripheral blood (PB)) IL-17A (T17) and/or interferon (IFN)-γ-producing (T1, T1/T17) T-cell populations in 86 pediatric controls and 116 age-matched CD patients upon phorbol myristate acetate/ionomycin or CD3/CD28 stimulation. T17 and T1/17 are physiologically present among IEL and PB populations, and their frequency is selectively and significantly reduced in CD IEL. The physiological age-dependent increase of Th17 IEL is also absent in CD, while IFN-γ-producing PB-T cells significantly accumulate with patient's age. Finally, the amplitude of IL-17A+ and IFN-γ+ T-cell pools are significantly correlated in different individuals; this relationship only applies to CD4+ T cells in controls, while it involves also the CD4- counterpart in CD patients. In conclusion, both size and dynamics of mucosa-associated and circulating IL-17A+ T-cell pools are finely regulated in human pediatric subjects, and severely disturbed in CD. The impaired IL-17A+ IEL-T pool may negatively impact on epithelial barrier efficiency, and contribute to CD mucosa damage; the disturbed dynamics of circulating IL-17A+ and IFN-γ+ T-cell pools may be involved in the extraintestinal autoimmune manifestations associated with CD.
Collapse
|
424
|
Lee SW, Park Y, Eun SY, Madireddi S, Cheroutre H, Croft M. Cutting edge: 4-1BB controls regulatory activity in dendritic cells through promoting optimal expression of retinal dehydrogenase. THE JOURNAL OF IMMUNOLOGY 2012; 189:2697-701. [PMID: 22896640 DOI: 10.4049/jimmunol.1201248] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Dendritic cells (DC) in the gut promote immune tolerance by expressing retinal dehydrogenase (RALDH), an enzyme that promotes retinoic acid, which aids differentiation of Foxp3+ inducible regulatory T cells (iTreg) in the intestinal mucosa. How RALDH expression is regulated is unclear. We found that 4-1BB (CD137), a member of the TNFR family, together with CD103, marked mesenteric lymph node DC with the highest level of RALDH activity, and ligation of 4-1BB maintained RALDH expression in these gut DC. Moreover, 4-1BB signals synergized with those through TLR2 or GM-CSFR to promote RALDH activity in undifferentiated DC. Correspondingly, 4-1BB-deficient mice were impaired in their ability to generate iTreg in the GALT when exposed to oral Ag, and 4-1BB-deficient mesenteric lymph node DC displayed weak RALDH activity and were poor at promoting iTreg development. Thus, our data demonstrate a novel activity of 4-1BB in controlling RALDH expression and the regulatory activity of DC.
Collapse
Affiliation(s)
- Seung-Woo Lee
- Division of Immune Regulation, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | | | | | | | | | | |
Collapse
|
425
|
Harley ITW, Karp CL. Obesity and the gut microbiome: Striving for causality. Mol Metab 2012; 1:21-31. [PMID: 24024115 DOI: 10.1016/j.molmet.2012.07.002] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2012] [Revised: 07/09/2012] [Accepted: 07/09/2012] [Indexed: 12/15/2022] Open
Abstract
The gut microbiome has been proposed to play a causal role in obesity. Here, we review the historical context for this hypothesis, highlight recent key findings, and critically discuss issues central to further progress in the field, including the central epistemological problem for the field: how to define causality in the relationship between microbiota and obesity phenotypes. Definition of such will be critical for the field to move forward.
Collapse
Affiliation(s)
- Isaac T W Harley
- Division of Molecular Immunology, Cincinnati Children's Hospital Research Foundation, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | | |
Collapse
|
426
|
Rendueles O, Ferrières L, Frétaud M, Bégaud E, Herbomel P, Levraud JP, Ghigo JM. A new zebrafish model of oro-intestinal pathogen colonization reveals a key role for adhesion in protection by probiotic bacteria. PLoS Pathog 2012; 8:e1002815. [PMID: 22911651 PMCID: PMC3406073 DOI: 10.1371/journal.ppat.1002815] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Accepted: 06/09/2012] [Indexed: 11/19/2022] Open
Abstract
The beneficial contribution of commensal bacteria to host health and homeostasis led to the concept that exogenous non-pathogenic bacteria called probiotics could be used to limit disease caused by pathogens. However, despite recent progress using gnotobiotic mammal and invertebrate models, mechanisms underlying protection afforded by commensal and probiotic bacteria against pathogens remain poorly understood. Here we developed a zebrafish model of controlled co-infection in which germ-free zebrafish raised on axenic living protozoa enabled the study of interactions between host and commensal and pathogenic bacteria. We screened enteric fish pathogens and identified Edwardsiella ictaluri as a virulent strain inducing a strong inflammatory response and rapid mortality in zebrafish larvae infected by the natural oro-intestinal route. Using mortality induced by infection as a phenotypic read-out, we pre-colonized zebrafish larvae with 37 potential probiotic bacterial strains and screened for survival upon E. ictaluri infection. We identified 3 robustly protective strains, including Vibrio parahaemolyticus and 2 Escherichia coli strains. We showed that the observed protective effect of E. coli was not correlated with a reduced host inflammatory response, nor with the release of biocidal molecules by protective bacteria, but rather with the presence of specific adhesion factors such as F pili that promote the emergence of probiotic bacteria in zebrafish larvae. Our study therefore provides new insights into the molecular events underlying the probiotic effect and constitutes a potentially high-throughput in vivo approach to the study of the molecular basis of pathogen exclusion in a relevant model of vertebrate oro-intestinal infection.
Collapse
Affiliation(s)
- Olaya Rendueles
- Institut Pasteur, Unité de Génétique des Biofilms, Département de Microbiologie, Paris, France
| | - Lionel Ferrières
- Institut Pasteur, Unité de Génétique des Biofilms, Département de Microbiologie, Paris, France
| | - Maxence Frétaud
- Institut Pasteur, Unité Macrophages et Développement de l'Immunité, Département de Biologie du Développement, Paris, France
- CNRS, URA2578, Paris, France
| | - Evelyne Bégaud
- Institut Pasteur, Centre de Ressources Biologiques de l'Institut Pasteur, Paris, France
| | - Philippe Herbomel
- Institut Pasteur, Unité Macrophages et Développement de l'Immunité, Département de Biologie du Développement, Paris, France
- CNRS, URA2578, Paris, France
| | - Jean-Pierre Levraud
- Institut Pasteur, Unité Macrophages et Développement de l'Immunité, Département de Biologie du Développement, Paris, France
- CNRS, URA2578, Paris, France
| | - Jean-Marc Ghigo
- Institut Pasteur, Unité de Génétique des Biofilms, Département de Microbiologie, Paris, France
- * E-mail:
| |
Collapse
|
427
|
Pott J, Hornef M. Innate immune signalling at the intestinal epithelium in homeostasis and disease. EMBO Rep 2012; 13:684-98. [PMID: 22801555 DOI: 10.1038/embor.2012.96] [Citation(s) in RCA: 144] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Accepted: 06/07/2012] [Indexed: 02/06/2023] Open
Abstract
The intestinal epithelium--which constitutes the interface between the enteric microbiota and host tissues--actively contributes to the maintenance of mucosal homeostasis and defends against pathogenic microbes. The recognition of conserved microbial products by cytosolic or transmembrane pattern recognition receptors in epithelial cells initiates signal transduction and influences effector cell function. However, the signalling pathways, effector molecules and regulatory mechanisms involved are not yet fully understood, and the functional outcome is poorly defined. This review analyses the complex and dynamic role of intestinal epithelial innate immune recognition and signalling, on the basis of results in intestinal epithelial cell-specific transgene or gene-deficient animals. This approach identifies specific epithelial cell functions within the diverse cellular composition of the mucosal tissue, in the presence of the complex and dynamic gut microbiota. These insights have thus provided a more comprehensive understanding of the role of the intestinal epithelium in innate immunity during homeostasis and disease.
Collapse
Affiliation(s)
- Johanna Pott
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK
| | | |
Collapse
|
428
|
Abstract
The 2011 Nobel Prize in Physiology/Medicine to Ralph Steinmann, Jules Hoffmann, and Bruce Beutler recognized a paradigm shift in our understanding of innate immunity, and its impact on adaptive immunity. The Prize highlighted the initial discoveries of Toll's role in immunity in flies, Toll-like receptors in mammals, and the establishment of dendritic cells as the initiators of adaptive immunity. This historical Commentary focuses on the developments in our understanding of innate immunity.
Collapse
Affiliation(s)
- Hermann Wagner
- Institute of Medicine Microbiology, Immunology and Hygiene, Technische Universität München, Trogerstr. 30, München, Germany.
| |
Collapse
|
429
|
Sullam KE, Essinger SD, Lozupone CA, O'Connor MP, Rosen GL, Knight R, Kilham SS, Russell JA. Environmental and ecological factors that shape the gut bacterial communities of fish: a meta-analysis. Mol Ecol 2012; 21:3363-78. [PMID: 22486918 PMCID: PMC3882143 DOI: 10.1111/j.1365-294x.2012.05552.x] [Citation(s) in RCA: 519] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Symbiotic bacteria often help their hosts acquire nutrients from their diet, showing trends of co-evolution and independent acquisition by hosts from the same trophic levels. While these trends hint at important roles for biotic factors, the effects of the abiotic environment on symbiotic community composition remain comparably understudied. In this investigation, we examined the influence of abiotic and biotic factors on the gut bacterial communities of fish from different taxa, trophic levels and habitats. Phylogenetic and statistical analyses of 25 16S rRNA libraries revealed that salinity, trophic level and possibly host phylogeny shape the composition of fish gut bacteria. When analysed alongside bacterial communities from other environments, fish gut communities typically clustered with gut communities from mammals and insects. Similar consideration of individual phylotypes (vs. communities) revealed evolutionary ties between fish gut microbes and symbionts of animals, as many of the bacteria from the guts of herbivorous fish were closely related to those from mammals. Our results indicate that fish harbour more specialized gut communities than previously recognized. They also highlight a trend of convergent acquisition of similar bacterial communities by fish and mammals, raising the possibility that fish were the first to evolve symbioses resembling those found among extant gut fermenting mammals.
Collapse
Affiliation(s)
- Karen E Sullam
- Department of Biology, Drexel University, Philadelphia, PA 19104, USA.
| | | | | | | | | | | | | | | |
Collapse
|
430
|
Neibergs HL, Johnson KA. ALPHARMA BEEF CATTLE NUTRITION SYMPOSIUM: Nutrition and the genome1. J Anim Sci 2012; 90:2308-16. [DOI: 10.2527/jas.2011-4582] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Affiliation(s)
- H. L. Neibergs
- Department of Animal Sciences, Washington State University, Pullman 99164
| | - K. A. Johnson
- Department of Animal Sciences, Washington State University, Pullman 99164
| |
Collapse
|
431
|
CCR7 deficiency causes diarrhea associated with altered ion transport in colonocytes in the absence of overt colitis. Mucosal Immunol 2012; 5:377-87. [PMID: 22395421 DOI: 10.1038/mi.2012.15] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The chemokine receptor CCR7 is a central regulator in the maintenance of cellular homeostasis of mucosal tissues. CCR7⁻/⁻ mice develop autoimmune gastritis and exocrinopathy accompanied by the formation of mucosal tertiary lymphoid follicles. Here we found that CCR7-deficient mice frequently suffered from chronic diarrhea linked with increased gastrointestinal motility and the development of severe anorectal prolapse. Enhanced formation of intestinal lymphoid follicles was associated with an elevated proportion of activated colonic T cells and increased production of the cytokine interleukin (IL)-1β. To uncover the pathomechanisms of diarrhea in CCR7⁻/⁻ mice, colonic epithelial barrier and ion channel activities were analyzed in Ussing chambers. Although overt acute colitis was absent, CCR7 deficiency resulted in reduced electrogenic sodium absorption and colonic chloride secretion. As it is known that IL-1β regulates epithelial sodium channel (ENaC) activity, these data imply a causal link between CCR7 expression, IL-1β level, and Na⁺ malabsorption owing to altered ENaC expression and diarrhea.
Collapse
|
432
|
Murine gut microbiota is defined by host genetics and modulates variation of metabolic traits. PLoS One 2012; 7:e39191. [PMID: 22723961 PMCID: PMC3377628 DOI: 10.1371/journal.pone.0039191] [Citation(s) in RCA: 168] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2011] [Accepted: 05/16/2012] [Indexed: 12/16/2022] Open
Abstract
The gastrointestinal tract harbors a complex and diverse microbiota that has an important role in host metabolism. Microbial diversity is influenced by a combination of environmental and host genetic factors and is associated with several polygenic diseases. In this study we combined next-generation sequencing, genetic mapping, and a set of physiological traits of the BXD mouse population to explore genetic factors that explain differences in gut microbiota and its impact on metabolic traits. Molecular profiling of the gut microbiota revealed important quantitative differences in microbial composition among BXD strains. These differences in gut microbial composition are influenced by host-genetics, which is complex and involves many loci. Linkage analysis defined Quantitative Trait Loci (QTLs) restricted to a particular taxon, branch or that influenced the variation of taxa across phyla. Gene expression within the gastrointestinal tract and sequence analysis of the parental genomes in the QTL regions uncovered candidate genes with potential to alter gut immunological profiles and impact the balance between gut microbial communities. A QTL region on Chr 4 that overlaps several interferon genes modulates the population of Bacteroides, and potentially Bacteroidetes and Firmicutes–the predominant BXD gut phyla. Irak4, a signaling molecule in the Toll-like receptor pathways is a candidate for the QTL on Chr15 that modulates Rikenellaceae, whereas Tgfb3, a cytokine modulating the barrier function of the intestine and tolerance to commensal bacteria, overlaps a QTL on Chr 12 that influence Prevotellaceae. Relationships between gut microflora, morphological and metabolic traits were uncovered, some potentially a result of common genetic sources of variation.
Collapse
|
433
|
Kiss EA, Diefenbach A. Role of the Aryl Hydrocarbon Receptor in Controlling Maintenance and Functional Programs of RORγt(+) Innate Lymphoid Cells and Intraepithelial Lymphocytes. Front Immunol 2012; 3:124. [PMID: 22666222 PMCID: PMC3364460 DOI: 10.3389/fimmu.2012.00124] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2012] [Accepted: 05/02/2012] [Indexed: 12/14/2022] Open
Abstract
Mucosal retinoic receptor-related orphan receptor (ROR)γt-expressing innate lymphoid cells (ILC) play an important role in the defense against intestinal pathogens and in promoting epithelial homeostasis and adaptation, thereby effectively protecting the vertebrate host against intestinal inflammatory disorders. The functional activity of RORγt(+) ILC is under the control of environmental cues. However, the molecular sensors for such environmental signals are largely unknown. Recently, the aryl hydrocarbon receptor (AhR) has emerged as a master regulator for the postnatal maintenance of intestinal RORγt(+) ILC and intraepithelial lymphocytes. AhR is a highly conserved transcription factor whose activity is regulated by environmental and dietary small molecule ligands. Here, we review the role of AhR signaling for the maintenance of intestinal immune cells and its impact on the immunological protection against intestinal infections and debilitating chronic inflammatory disorders.
Collapse
Affiliation(s)
- Elina A Kiss
- Institute of Medical Microbiology and Hygiene, University of Freiburg Freiburg, Germany
| | | |
Collapse
|
434
|
Hernández-Hernández O, Marín-Manzano MC, Rubio LA, Moreno FJ, Sanz ML, Clemente A. Monomer and Linkage Type of Galacto-Oligosaccharides Affect Their Resistance to Ileal Digestion and Prebiotic Properties in Rats. J Nutr 2012; 142:1232-9. [DOI: 10.3945/jn.111.155762] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Affiliation(s)
| | - M. Carmen Marín-Manzano
- Estación Experimental del Zaidín, Consejo Superior de Investigaciones Científicas, Granada, Spain
| | - Luis A. Rubio
- Estación Experimental del Zaidín, Consejo Superior de Investigaciones Científicas, Granada, Spain
| | - F. Javier Moreno
- Instituto de Investigación en Ciencias de la Alimentación, Campus de Cantoblanco, Universidad Autónoma de Madrid-Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - M. Luz Sanz
- Instituto de Química Orgánica General, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Alfonso Clemente
- Estación Experimental del Zaidín, Consejo Superior de Investigaciones Científicas, Granada, Spain
| |
Collapse
|
435
|
Bernardo D, Sánchez B, Al-Hassi HO, Mann ER, Urdaci MC, Knight SC, Margolles A. Microbiota/host crosstalk biomarkers: regulatory response of human intestinal dendritic cells exposed to Lactobacillus extracellular encrypted peptide. PLoS One 2012; 7:e36262. [PMID: 22606249 PMCID: PMC3351486 DOI: 10.1371/journal.pone.0036262] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2012] [Accepted: 04/04/2012] [Indexed: 02/07/2023] Open
Abstract
The human gastrointestinal tract is exposed to a huge variety of microorganisms, either commensal or pathogenic; at this site, a balance between immunity and immune tolerance is required. Intestinal dendritic cells (DCs) control the mechanisms of immune response/tolerance in the gut. In this paper we have identified a peptide (STp) secreted by Lactobacillus plantarum, characterized by the abundance of serine and threonine residues within its sequence. STp is encoded in one of the main extracellular proteins produced by such species, which includes some probiotic strains, and lacks cleavage sites for the major intestinal proteases. When studied in vitro, STp expanded the ongoing production of regulatory IL-10 in human intestinal DCs from healthy controls. STp-primed DC induced an immunoregulatory cytokine profile and skin-homing profile on stimulated T-cells. Our data suggest that some of the molecular dialogue between intestinal bacteria and DCs may be mediated by immunomodulatory peptides, encoded in larger extracellular proteins, secreted by commensal bacteria. These peptides may be used for the development of nutraceutical products for patients with IBD. In addition, this kind of peptides seem to be absent in the gut of inflammatory bowel disease patients, suggesting a potential role as biomarker of gut homeostasis.
Collapse
Affiliation(s)
- David Bernardo
- Antigen Presentation Research Group, Imperial College London, Harrow, United Kingdom
| | - Borja Sánchez
- Departmento de Microbiología y Bioquímica de Productos Lácteos, Instituto de Productos Lácteos de Asturias, Consejo Superior de Investigaciones Científicas, Villaviciosa, Spain
| | - Hafid O. Al-Hassi
- Antigen Presentation Research Group, Imperial College London, Harrow, United Kingdom
| | - Elizabeth R. Mann
- Antigen Presentation Research Group, Imperial College London, Harrow, United Kingdom
| | - María C. Urdaci
- Laboratoire de Microbiologie et Biochimie Appliquée, Ecole Nationale Supérieure des Sciences Agronomiques de Bordeaux, Gradignan, France
| | - Stella C. Knight
- Antigen Presentation Research Group, Imperial College London, Harrow, United Kingdom
| | - Abelardo Margolles
- Departmento de Microbiología y Bioquímica de Productos Lácteos, Instituto de Productos Lácteos de Asturias, Consejo Superior de Investigaciones Científicas, Villaviciosa, Spain
| |
Collapse
|
436
|
Gomez A, Luckey D, Yeoman CJ, Marietta EV, Berg Miller ME, Murray JA, White BA, Taneja V. Loss of sex and age driven differences in the gut microbiome characterize arthritis-susceptible 0401 mice but not arthritis-resistant 0402 mice. PLoS One 2012; 7:e36095. [PMID: 22553482 PMCID: PMC3338357 DOI: 10.1371/journal.pone.0036095] [Citation(s) in RCA: 178] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2011] [Accepted: 03/27/2012] [Indexed: 11/24/2022] Open
Abstract
Background HLA-DRB1*0401 is associated with susceptibility, while HLA-DRB1*0402 is associated with resistance to developing rheumatoid arthritis (RA) and collagen-induced arthritis in humans and transgenic mice respectively. The influence of gut-joint axis has been suggested in RA, though not yet proven. Methodology/Principal Findings We have used HLA transgenic mice carrying arthritis susceptible and -resistant HLA-DR genes to explore if genetic factors and their interaction with gut flora gut can be used to predict susceptibility to develop arthritis. Pyrosequencing of the 16S rRNA gene from the fecal microbiomes of DRB1*0401 and DRB1*0402 transgenic mice revealed that the guts of *0401 mice is dominated by a Clostridium-like bacterium, whereas the guts of *0402 mice are enriched for members of the Porphyromonadaceae family and Bifidobacteria. DRB1*0402 mice harbor a dynamic sex and age-influenced gut microbiome while DRB1*0401 mice did not show age and sex differences in gut microbiome even though they had altered gut permeability. Cytokine transcripts, measured by rtPCR, in jejuna showed differential TH17 regulatory network gene transcripts in *0401 and *0402 mice. Conclusions/Significance We have demonstrated for the first time that HLA genes in association with the gut microbiome may determine the immune environment and that the gut microbiome might be a potential biomarker as well as contributor for susceptibility to arthritis. Identification of pathogenic commensal bacteria would provide new understanding of disease pathogenesis, thereby leading to novel approaches for therapy.
Collapse
Affiliation(s)
- Andres Gomez
- Institute for Genomic Biology, University of Illinois, Urbana, Illinois, United States of America
| | - David Luckey
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Carl J. Yeoman
- Institute for Genomic Biology, University of Illinois, Urbana, Illinois, United States of America
| | - Eric V. Marietta
- Department of Gasteroenterology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Margret E. Berg Miller
- Institute for Genomic Biology, University of Illinois, Urbana, Illinois, United States of America
| | - Joseph A. Murray
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, United States of America
- Department of Gasteroenterology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Bryan A. White
- Institute for Genomic Biology, University of Illinois, Urbana, Illinois, United States of America
- Department of Animal Sciences, University of Illinois, Urbana, Illinois, United States of America
- * E-mail: (BAW); (VT)
| | - Veena Taneja
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, United States of America
- Department of Rheumatology, Mayo Clinic, Rochester, Minnesota, United States of America
- * E-mail: (BAW); (VT)
| |
Collapse
|
437
|
Kelly D, Delday MI, Mulder I. Microbes and microbial effector molecules in treatment of inflammatory disorders. Immunol Rev 2012; 245:27-44. [PMID: 22168412 DOI: 10.1111/j.1600-065x.2011.01079.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The healthy gut tolerates very large numbers of diverse bacterial species belonging mainly to the Bacteroidetes and Firmicutes phyla. These bacteria normally coexist peacefully with the gut and help maintain immune homeostasis and tolerance. The mechanisms promoting tolerance affect various cell populations, including the epithelial cells lining the gut, resident dendritic cells (DCs), and gut-homing T cells. Gut bacteria also influence multiple signaling pathways from Toll-like receptors to nuclear factor κB and regulate the functionality of DCs and T cells. Several bacterial species have been identified that promote T-cell differentiation, in particular T-helper 17 and T-regulatory cells. Insight into the molecular mechanisms by which bacteria mediate these effects will be very important in identifying new ways of treating intestinal and extra-intestinal immune-mediated diseases. These diseases are increasing dramatically in the human population and require new treatments. It may be possible in the future to identify specific bacterial species or strains that can correct for T-cell imbalances in the gut and promote immune homeostasis, both locally and systemically. In addition, new information describing microbial genomes affords the opportunity to mine for functional genes that may lead to new generation drugs relevant to a range of inflammatory disease conditions.
Collapse
Affiliation(s)
- Denise Kelly
- Gut Immunology Group, Rowett Institute of Nutrition and Health, University of Aberdeen, Aberdeen, UK.
| | | | | |
Collapse
|
438
|
Rodriguez B, Prioult G, Hacini-Rachinel F, Moine D, Bruttin A, Ngom-Bru C, Labellie C, Nicolis I, Berger B, Mercenier A, Butel MJ, Waligora-Dupriet AJ. Infant gut microbiota is protective against cow's milk allergy in mice despite immature ileal T-cell response. FEMS Microbiol Ecol 2012; 79:192-202. [PMID: 22029421 DOI: 10.1111/j.1574-6941.2011.01207.x] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Faecal microbiota of healthy infant displays a large abundance of Bifidobacterium spp. and Bacteroides spp. Although some studies have reported an association between these two genera and allergy, these findings remain a subject of debate. Using a gnotobiotic mouse model of cow's milk allergy, we investigated the impact of an infant gut microbiota – mainly composed of Bifidobacterium and Bacteroides spp. – on immune activation and allergic manifestations. The transplanted microbiota failed to restore an ileal T-cell response similar to the one observed in conventional mice. This may be due to the low bacterial translocation into Peyer's patches in gnotobiotic mice. The allergic response was then monitored in germ-free, gnotobiotic, and conventional mice after repeated oral sensitization with whey proteins and cholera toxin. Colonized mice displayed a lower drop of rectal temperature upon oral challenge with b-lactoglobulin, lower plasma mMCP-1, and lower anti-BLG IgG1 than germ-free mice. The foxp3 gene was highly expressed in the ileum of both colonized mice that were protected against allergy. This study is the first demonstration that a transplanted healthy infant microbiota mainly composed of Bifidobacterium and Bacteroides had a protective impact on sensitization and food allergy in mice despite altered T-cell response in the ileum.
Collapse
Affiliation(s)
- Bertrand Rodriguez
- Facultédes Sciences Pharmaceutiques et Biologiques, EA 4065, Département Périnatalité, Microbiologie, Médicament, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
439
|
Chervonsky AV. Intestinal commensals: influence on immune system and tolerance to pathogens. Curr Opin Immunol 2012; 24:255-60. [PMID: 22445718 DOI: 10.1016/j.coi.2012.03.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Accepted: 03/04/2012] [Indexed: 11/28/2022]
Abstract
The interactions of commensal microorganisms with the host's immune system are in the spotlight. The intestinal microbiota provides both stimulatory and inhibitory signals to the host ensuring its own survival and contributing to resistance to pathogens. Some microbial lineages do this better than others and are attracting a lot of attention. The microbial influences go beyond the gut and have profound effects on infections and autoimmunity in distant locations. Commensals are also involved in regulation of 'tolerance to pathogens', a fundamental type of response to infections that does not reduce pathogen burden but keeps the host healthy.
Collapse
Affiliation(s)
- Alexander V Chervonsky
- Department of Pathology, University of Chicago, 924 57th Street, Chicago, IL 60637, United States.
| |
Collapse
|
440
|
Vitetta L, Briskey D, Hayes E, Shing C, Peake J. A review of the pharmacobiotic regulation of gastrointestinal inflammation by probiotics, commensal bacteria and prebiotics. Inflammopharmacology 2012; 20:251-66. [PMID: 22427210 DOI: 10.1007/s10787-012-0126-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2012] [Accepted: 02/07/2012] [Indexed: 12/17/2022]
Abstract
The idea that microbes induce disease has steered medical research toward the discovery of antibacterial products for the prevention and treatment of microbial infections. The twentieth century saw increasing dependency on antimicrobials as mainline therapy accentuating the notion that bacterial interactions with humans were to be avoided or desirably controlled. The last two decades, though, have seen a refocusing of thinking and research effort directed towards elucidating the critical inter-relationships between the gut microbiome and its host that control health/wellness or disease. This research has redefined the interactions between gut microbes and vertebrates, now recognizing that the microbial active cohort and its mammalian host have shared co-evolutionary metabolic interactions that span millennia. Microbial interactions in the gastrointestinal tract provide the necessary cues for the development of regulated pro- and anti-inflammatory signals that promotes immunological tolerance, metabolic regulation and other factors which may then control local and extra-intestinal inflammation. Pharmacobiotics, using nutritional and functional food additives to regulate the gut microbiome, will be an exciting growth area of therapeutics, developing alongside an increased scientific understanding of gut-microbiome symbiosis in health and disease.
Collapse
Affiliation(s)
- L Vitetta
- School of Medicine, Centre for Integrative Clinical and Molecular Medicine, Princess Alexandra Hospital, The University of Queensland, Lvl 2, R Wing, 199 Ipswich Road, Woolloongabba, Brisbane, QLD 4102, Australia.
| | | | | | | | | |
Collapse
|
441
|
De Cruz P, Prideaux L, Wagner J, Ng SC, McSweeney C, Kirkwood C, Morrison M, Kamm MA. Characterization of the gastrointestinal microbiota in health and inflammatory bowel disease. Inflamm Bowel Dis 2012; 18:372-90. [PMID: 21604329 DOI: 10.1002/ibd.21751] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2011] [Accepted: 03/31/2011] [Indexed: 02/06/2023]
Abstract
The enteric bacterial flora play a key role in maintaining health. Inflammatory bowel disease is associated with quantitative and qualitative alterations in the microbiota. Early characterization of the microbiota involved culture-dependent techniques. The advent of metagenomic techniques, however, allows for structural and functional characterization using culture-independent methods. Changes in diversity, together with quantitative alterations in specific bacterial species, have been identified. The functional significance of these changes, and their pathogenic role, remain to be elucidated.
Collapse
|
442
|
Abstract
Over the last decades the rising occurrence of metabolic diseases throughout the world points to the failure of preventive and therapeutic strategies and of the corresponding molecular and physiological concepts. Therefore, a new paradigm needs to be elucidated. Very recently the intimate cross talk of the intestinal microbiota with the host immune system has opened new avenues. The large diversity of the intestinal microbes' genome, i.e. the metagenome, and the extreme plasticity of the immune system provide a unique balance which, when finely tuned, maintains a steady homeostasis. The discovery that a new microbiota repertoire is one of the causes responsible for the onset of metabolic disease suggests that the relationship with the immune system is impaired. Therefore, we here review the recent arguments that support the view that an alteration in the microbiota to host immune system balance leads to an increased translocation of bacterial antigens towards metabolically active tissues, and could result in a chronic inflammatory state and consequently impaired metabolic functions such as insulin resistance, hepatic fat deposition, insulin unresponsiveness, and excessive adipose tissue development. This imbalance could be at the onset of metabolic disease, and therefore the early treatment of the microbiota dysbiosis or immunomodulatory strategies should prevent and slow down the epidemic of metabolic diseases and hence the corresponding lethal cardiovascular consequences.
Collapse
Affiliation(s)
- Rémy Burcelin
- Institut National de la Santé et de la Recherche Médicale, U1048, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), BP 84225, 31432 Toulouse, France.
| | | | | |
Collapse
|
443
|
Wekerle H, Flügel A, Fugger L, Schett G, Serreze D. Autoimmunity's next top models. Nat Med 2012; 18:66-70. [PMID: 22227675 DOI: 10.1038/nm.2635] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Hartmut Wekerle
- Department of Neuroimmunology, Max Planck Institute of Neurobiology, Munich, Germany.
| | | | | | | | | |
Collapse
|
444
|
Serum sTREM-1 as a surrogate marker of treatment outcome in patients with peptic ulcer disease. Dig Dis Sci 2011; 56:3590-5. [PMID: 21633832 DOI: 10.1007/s10620-011-1761-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2011] [Accepted: 05/14/2011] [Indexed: 12/09/2022]
Abstract
OBJECTIVE Soluble triggering receptor expressed on myeloid cells-1 (sTREM-1) is elevated in the gastric juice and in cultures of gastric mucosa of patients with peptic ulcer disease (PUD). Its application as a surrogate marker for the treatment of PUD was assessed. METHODS From 138 eligible patients, 96 were enrolled; 50 with duodenal ulcer, 29 with gastric ulcer and 17 with chronic gastritis. Patients were endoscoped twice; once before treatment and once after treatment. Biopsy specimens were collected for histopathologic estimation of gastritis. Blood was sampled prior to each endoscopy. Serum was collected and sTREM-1 was measured by an enzyme immunoabsorbent assay ( http://www.clinicaltrials.gov identifier NCT00534443). RESULTS At the end of treatment sTREM-1 was either: (a) below the limit of detection (this occurred in 62 patients and it was accompanied by lacks signs of residual disease in 58 patients, 93.5%); or (b) above the limit of detection (this occurred in 17 patients and it was accompanied by residual disease in 14 patients, 82.3%) (p < 0.0001). Odds ratio for complete healing of peptic ulcer with sTREM-1 below detection limit was 5.30 (95% CI: 1.89-14.83, p < 0.001) compared to serum sTREM-1 above the limit of detection. CONCLUSIONS Serum sTREM-1 below detection limit may effectively distinguish patients who successfully completed therapy for PUD from those with residual disease and apply as a surrogate marker.
Collapse
|
445
|
Crosstalk between B lymphocytes, microbiota and the intestinal epithelium governs immunity versus metabolism in the gut. Nat Med 2011; 17:1585-93. [PMID: 22101768 DOI: 10.1038/nm.2505] [Citation(s) in RCA: 271] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2011] [Accepted: 09/09/2011] [Indexed: 12/13/2022]
Abstract
Using a systems biology approach, we discovered and dissected a three-way interaction between the immune system, the intestinal epithelium and the microbiota. We found that, in the absence of B cells, or of IgA, and in the presence of the microbiota, the intestinal epithelium launches its own protective mechanisms, upregulating interferon-inducible immune response pathways and simultaneously repressing Gata4-related metabolic functions. This shift in intestinal function leads to lipid malabsorption and decreased deposition of body fat. Network analysis revealed the presence of two interconnected epithelial-cell gene networks, one governing lipid metabolism and another regulating immunity, that were inversely expressed. Gene expression patterns in gut biopsies from individuals with common variable immunodeficiency or with HIV infection and intestinal malabsorption were very similar to those of the B cell-deficient mice, providing a possible explanation for a longstanding enigmatic association between immunodeficiency and defective lipid absorption in humans.
Collapse
|
446
|
Abstract
This review examines mechanisms by which the bacteria present in the gut interact with nutrients and host biology to affect the risk of obesity and associated disorders, including diabetes, inflammation, and liver diseases. The bacterial metabolism of nutrients in the gut is able to drive the release of bioactive compounds (including short-chain fatty acids or lipid metabolites), which interact with host cellular targets to control energy metabolism and immunity. Animal and human data demonstrate that phylogenic changes occur in the microbiota composition in obese versus lean individuals; they suggest that the count of specific bacteria is inversely related to fat mass development, diabetes, and/or the low levels of inflammation associated with obesity. The prebiotic and probiotic approaches are presented as interesting research tools to counteract the drop in target bacteria and thereby to estimate their relevance in the improvement of host metabolism.
Collapse
Affiliation(s)
- Nathalie M Delzenne
- Louvain Drug Research Institute, Metabolism and Nutrition Research Group, Université catholique de Louvain, Brussels, Belgium.
| | | |
Collapse
|
447
|
Peptidoglycan recognition proteins: modulators of the microbiome and inflammation. Nat Rev Immunol 2011; 11:837-51. [DOI: 10.1038/nri3089] [Citation(s) in RCA: 259] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
448
|
Swiatczak B, Rescigno M, Cohen IR. Systemic features of immune recognition in the gut. Microbes Infect 2011; 13:983-91. [DOI: 10.1016/j.micinf.2011.06.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2011] [Revised: 06/23/2011] [Accepted: 06/24/2011] [Indexed: 12/15/2022]
|
449
|
Edwards LA, Bajaj-Elliott M, Klein NJ, Murch SH, Phillips AD. Bacterial-epithelial contact is a key determinant of host innate immune responses to enteropathogenic and enteroaggregative Escherichia coli. PLoS One 2011; 6:e27030. [PMID: 22046438 PMCID: PMC3203933 DOI: 10.1371/journal.pone.0027030] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2011] [Accepted: 10/09/2011] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Enteropathogenic (EPEC) and Enteroaggregative (EAEC) E. coli have similar, but distinct clinical symptoms and modes of pathogenesis. Nevertheless when they infect the gastrointestinal tract, it is thought that their flagellin causes IL-8 release leading to neutrophil recruitment and gastroenteritis. However, this may not be the whole story as the effect of bacterial adherence to IEC innate response(s) remains unclear. Therefore, we have characterized which bacterial motifs contribute to the innate epithelial response to EPEC and EAEC, using a range of EPEC and EAEC isogenic mutant strains. METHODOLOGY Caco-2 and HEp-2 cell lines were exposed to prototypical EPEC strain E2348/69 or EAEC strain O42, in addition to a range of isogenic mutant strains. E69 [LPS, non-motile, non-adherent, type three secretion system (TTSS) negative, signalling negative] or O42 [non-motile, non-adherent]. IL-8 and CCL20 protein secretion was measured. Bacterial surface structures were assessed by negative staining Transmission Electron Microscopy. The Fluorescent-actin staining test was carried out to determine bacterial adherence. RESULTS Previous studies have reported a balance between the host pro-inflammatory response and microbial suppression of this response. In our system an overall balance towards the host pro-inflammatory response is seen with the E69 WT and to a greater extent O42 WT, which is in fit with clinical symptoms. On removal of the external EPEC structures flagella, LPS, BFP, EspA and EspC; and EAEC flagella and AAF, the host inflammatory response is reduced. However, removal of E69 lymphostatin increases the host inflammatory response suggesting involvement in the bacterial mediated anti-inflammatory response. CONCLUSION Epithelial responses were due to combinations of bacterial agonists, with host-bacterial contact a key determinant of these innate responses. Host epithelial recognition was offset by the microbe's ability to down-regulate the inflammatory response. Understanding the complexity of this host-microbial balance will contribute to improved vaccine design for infectious gastroenteritis.
Collapse
Affiliation(s)
- Lindsey A Edwards
- Centre for Paediatric Gastroenterology, Royal Free Hospital, London, United Kingdom.
| | | | | | | | | |
Collapse
|
450
|
Van Hauwermeiren F, Vandenbroucke RE, Libert C. Treatment of TNF mediated diseases by selective inhibition of soluble TNF or TNFR1. Cytokine Growth Factor Rev 2011; 22:311-9. [PMID: 21962830 DOI: 10.1016/j.cytogfr.2011.09.004] [Citation(s) in RCA: 96] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The TNF signaling pathway is a valuable target in the therapy of autoimmune diseases, and anti-TNF drugs are successfully used to treat diseases such as rheumatoid arthritis, Crohn's disease and psoriasis. By their ability to interfere with inflammatory processes at multiple levels, these TNF blockers have become invaluable tools to inhibit the inflammation induced damage and allow recovery of the affected tissues. Unfortunately this therapy has some drawbacks, including increased risk of infection and malignancy, and remarkably, the onset of new auto-immune diseases. Some of these effects are caused by the unwanted abrogation of beneficial TNF signaling. More specific targeting of the pathological TNF-induced signaling might lead to broader applicability and improved safety. Specificity might be increased by inhibiting the soluble TNF/TNFR1 axis while leaving the often beneficial transmembrane TNF/TNFR2 signaling untouched. This approach looks promising because it inhibits the pathological effects of TNF and reduces the side effects, and it opens the way for the treatment of other diseases in which TNFR2 inhibition is detrimental. In this review we give an overview of in vivo mouse studies of TNF mediated pathologies demonstrating that the blockade or genetic deletion of sTNF or TNFR1 is preferable over total TNF blockade.
Collapse
|