51
|
Pectin Interaction with Immune Receptors is Modulated by Ripening Process in Papayas. Sci Rep 2020; 10:1690. [PMID: 32015377 PMCID: PMC6997392 DOI: 10.1038/s41598-020-58311-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 01/07/2020] [Indexed: 01/05/2023] Open
Abstract
Dietary fibers have been shown to exert immune effects via interaction with pattern recognition receptors (PRR) such as toll-like receptors (TLR) and nucleotide-binding oligomerization domain (NOD)-like receptors. Pectin is a dietary fiber that interacts with PRR depending on its chemical structure. Papaya pectin retains different chemical structures at different ripening stages. How this influence PRR signaling is unknown. The aim of this work was to determine how ripening influences pectin structures and their ability to interact with TLR2, 3, 4, 5 and 9, and NOD1 and 2. It was evaluated the interaction of the water-soluble fractions rich in pectin extracted from unripe to ripe papayas. The pectin extracted from ripe papayas activated all the TLR and, to a lesser extent, the NOD receptors. The pectin extracted from unripe papayas also activated TLR2, 4 and 5 but inhibited the activation of TLR3 and 9. The differences in pectin structures are the higher methyl esterification and smaller galacturonan chains of pectin from ripe papayas. Our finding might lead to selection of ripening stages for tailored modulation of PRR to support or attenuate immunity.
Collapse
|
52
|
Song J, Li Q, Everaert N, Liu R, Zheng M, Zhao G, Wen J. Effects of inulin supplementation on intestinal barrier function and immunity in specific pathogen-free chickens with Salmonella infection. J Anim Sci 2020; 98:skz396. [PMID: 31894241 PMCID: PMC6986778 DOI: 10.1093/jas/skz396] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 12/30/2019] [Indexed: 12/11/2022] Open
Abstract
We investigated the effects of inulin on intestinal barrier function and mucosal immunity in Salmonella enterica serovar Enteritidis (SE)-infected specific pathogen-free (SPF) chickens. SPF chickens (n = 240, 1-d-old) were divided into 4 groups (6 replicates per group, 10 chickens per replicate): a control group (CON) fed a basal diet without inulin supplementation and 3 SE-infected groups fed a basal diet supplemented with inulin 0% (SE group), 0.5% (0.5% InSE group), and 1% (1% InSE group), respectively. At 28 d of age, the chickens in SE-infected groups were orally infected with SE and in CON group were administrated with phosphated-buffered saline (PBS). Intestinal morphology, mucosal immunity, and intestinal barrier function-related gene expression were analyzed at 1- and 3-d post-infection (dpi). SE challenge significantly increased the mucosal gene expression, such as interleukin-1β (IL-1β), lipopolysaccharide-induced tumor necrosis factor factor (LITAF), interferon-γ (IFN-γ), and interleukin-6 (IL-6), and increased serum IFN-γ, secretory IgA (sIgA), and IgG concentration, and significantly decreased the gene expression levels of mucin 2 (MUC2) and claudin-1 at 3 dpi compared with the CON group (P < 0.05). Inulin supplementation improved the expression levels of these immunity- and intestinal barrier function-related genes, increased villus height (VH), and decreased crypt depth (CD) in the duodenum, jejunum, and ileum at 1 and 3 dpi within the SE-challenged groups (P < 0.05). SE challenge significantly increased ileal Toll-like receptor 4 (TLR4) mRNA at 1 and 3 dpi, suppressor of cytokine signaling 3 (SOCS3) mRNA at 1 dpi, and phospho-signal transducer and activator of transcription 3 (p-STAT3) and Janus kinase1 (JAK1) protein expression at 3 dpi compared with the CON group (P < 0.05). Inulin supplementation suppressed p-STAT3 and JAK1 protein expression and promoted ileal TLR4 and SOCS3 mRNA expression at 3 dpi compared with SE group (P < 0.05). In conclusion, inulin alleviated SE-induced gut injury by decreasing the proinflammatory response and enhancing mucosal immunity in chickens.
Collapse
Affiliation(s)
- Jiao Song
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, People’s Republic of China
| | - Qinghe Li
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, People’s Republic of China
| | - Nadia Everaert
- Precision Livestock and Nutrition Unit, Gembloux Agro-Bio Tech, University of Liège, Gembloux, Belgium
| | - Ranran Liu
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, People’s Republic of China
| | - Maiqing Zheng
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, People’s Republic of China
| | - Guiping Zhao
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, People’s Republic of China
| | - Jie Wen
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, People’s Republic of China
| |
Collapse
|
53
|
Cai Y, Folkerts J, Folkerts G, Maurer M, Braber S. Microbiota-dependent and -independent effects of dietary fibre on human health. Br J Pharmacol 2019; 177:1363-1381. [PMID: 31663129 DOI: 10.1111/bph.14871] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 09/06/2019] [Accepted: 09/08/2019] [Indexed: 12/11/2022] Open
Abstract
Dietary fibre, such as indigestible oligosaccharides and polysaccharides, occurs in many foods and has gained considerable importance related to its beneficial effects on host health and specific diseases. Dietary fibre is neither digested nor absorbed in the small intestine and modulates the composition of the gut microbiota. New evidence indicates that dietary fibre also interacts directly with the epithelium and immune cells throughout the gastrointestinal tract by microbiota-independent effects. This review focuses on how dietary fibre improves human health and the reported health benefits that are connected to molecular pathways, in (a) a microbiota-independent manner, via interaction with specific surface receptors on epithelial and immune cells regulating intestinal barrier and immune function, and (b) a microbiota-dependent manner via maintaining intestinal homeostasis by promoting beneficial microbes, including Bifidobacteria and Lactobacilli, limiting the growth, adhesion, and cytotoxicity of pathogenic microbes, as well as stimulating fibre-derived microbial short-chain fatty acid production. LINKED ARTICLES: This article is part of a themed section on The Pharmacology of Nutraceuticals. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v177.6/issuetoc.
Collapse
Affiliation(s)
- Yang Cai
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Jelle Folkerts
- Department of Pulmonary Medicine, Erasmus MC, Rotterdam, Netherlands.,Dermatological Allergology, Allergie-Centrum-Charité, Department of Dermatology and Allergy, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Gert Folkerts
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Marcus Maurer
- Dermatological Allergology, Allergie-Centrum-Charité, Department of Dermatology and Allergy, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Saskia Braber
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
54
|
Dendrobium huoshanense polysaccharide regulates intestinal lamina propria immune response by stimulation of intestinal epithelial cells via toll-like receptor 4. Carbohydr Polym 2019; 222:115028. [DOI: 10.1016/j.carbpol.2019.115028] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 06/16/2019] [Accepted: 06/25/2019] [Indexed: 12/14/2022]
|
55
|
Prebiotics: Mechanisms and Preventive Effects in Allergy. Nutrients 2019; 11:nu11081841. [PMID: 31398959 PMCID: PMC6722770 DOI: 10.3390/nu11081841] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 07/31/2019] [Accepted: 08/01/2019] [Indexed: 12/13/2022] Open
Abstract
Allergic diseases now affect over 30% of individuals in many communities, particularly young children, underscoring the need for effective prevention strategies in early life. These allergic conditions have been linked to environmental and lifestyle changes driving the dysfunction of three interdependent biological systems: microbiota, epithelial barrier and immune system. While this is multifactorial, dietary changes are of particular interest in the altered establishment and maturation of the microbiome, including the associated profile of metabolites that modulate immune development and barrier function. Prebiotics are non-digestible food ingredients that beneficially influence the health of the host by 1) acting as a fermentable substrate for some specific commensal host bacteria leading to the release of short-chain fatty acids in the gut intestinal tract influencing many molecular and cellular processes; 2) acting directly on several compartments and specifically on different patterns of cells (epithelial and immune cells). Nutrients with prebiotic properties are therefore of central interest in allergy prevention for their potential to promote a more tolerogenic environment through these multiple pathways. Both observational studies and experimental models lend further credence to this hypothesis. In this review, we describe both the mechanisms and the therapeutic evidence from preclinical and clinical studies exploring the role of prebiotics in allergy prevention.
Collapse
|
56
|
Rhein protects against barrier disruption and inhibits inflammation in intestinal epithelial cells. Int Immunopharmacol 2019; 71:321-327. [PMID: 30952096 DOI: 10.1016/j.intimp.2019.03.030] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 03/14/2019] [Accepted: 03/17/2019] [Indexed: 12/27/2022]
Abstract
BACKGROUND AND AIMS Intestinal epithelial barrier and intestinal inflammation play indispensable roles in the development of intestinal diseases. The major aims of the current study were to investigate the potential of rhein, a major flavonoid compound isolated from Rheum rhabarbarum, in the treatment of intestinal diseases and its underlying mechanisms in vitro. METHODS The protective role of rhein on intestinal epithelial barrier was evaluated in a monolayer of IEC-6 cells stimulated by TNF-α, while the anti-inflammatory effects were investigated in an IEC-6 cell model with LPS stimulation. RESULTS Rhein inhibited the increase of phenol red flux and the decrease of TEER, as well as recovered the expression and distribution of ZO-1 and weakened MLC phosphorylation, MLCK expression and NF-κB activation. Meanwhile, LPS-stimulated IL-1β and IL-6 were down-regulated, expression levels of TLR4, NLRP3 and cleaved caspase1 were weakened and NF-κB was inactivated. CONCLUSIONS These results suggested that rhein has potential therapeutic effects against intestinal diseases by maintaining intestinal epithelial barrier and suppressing intestinal inflammation.
Collapse
|
57
|
Castro-Alves VC, Shiga TM, Nascimento JROD. Polysaccharides from chayote enhance lipid efflux and regulate NLRP3 inflammasome priming in macrophage-like THP-1 cells exposed to cholesterol crystals. Int J Biol Macromol 2019; 127:502-510. [PMID: 30658148 DOI: 10.1016/j.ijbiomac.2019.01.048] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 12/24/2018] [Accepted: 01/10/2019] [Indexed: 01/14/2023]
Abstract
The contribution of dietary fiber to decrease the risk of atherosclerosis may occur through other mechanisms besides the increased excretion of cholesterol. Although macrophages are crucial for lipid clearance, the excessive uptake of cholesterol crystals (CC) by these cells induce NLRP3 inflammasome and foam cell formation. Thus, we investigated whether the water-soluble DF from chayote (WSP) regulate CC-pretreated macrophage-like THP-1 cells. Linkage analysis indicated that WSP is composed mainly of pectic homogalacturonan and highly branched type I rhamnogalacturonan as well as hemicellulosic material including glucomannan, xyloglucan, and glucurono(arabino)xylan. WSP reduced interleukin (IL)-1β and chemokine release in CC-pretreated macrophages. Notably, WSP also reduced lipid accumulation in cells previously exposed to CC. Furthermore, WSP upregulated liver X receptor alpha expression, which may account for increased lipid efflux, and reduced matrix metallopeptidase 9 expression. WSP also reduced active caspase-1 protein levels, and downregulated NLRP3 and IL-1β gene expression in CC-pretreated cells, suggesting that this polysaccharide fraction regulates the priming signals required for NLRP3 inflammasome activation. Thus, WSP regulate lipid efflux and suppress inflammasome priming in macrophages, suggesting that the health benefits of this dietary fiber could go beyond its physical properties on the gastrointestinal tract.
Collapse
Affiliation(s)
- Victor Costa Castro-Alves
- Department of Food Science and Experimental Nutrition, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, SP, Brazil; Food Research Center (FoRC), CEPID-FAPESP (Research, Innovation and Dissemination Centers, São Paulo Research Foundation), São Paulo, Brazil
| | - Tânia Misuzu Shiga
- Department of Food Science and Experimental Nutrition, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, SP, Brazil; Food Research Center (FoRC), CEPID-FAPESP (Research, Innovation and Dissemination Centers, São Paulo Research Foundation), São Paulo, Brazil
| | - João Roberto Oliveira do Nascimento
- Department of Food Science and Experimental Nutrition, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, SP, Brazil; Food Research Center (FoRC), CEPID-FAPESP (Research, Innovation and Dissemination Centers, São Paulo Research Foundation), São Paulo, Brazil; Food and Nutrition Research Center (NAPAN), University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
58
|
Anzola A, González R, Gámez-Belmonte R, Ocón B, Aranda CJ, Martínez-Moya P, López-Posadas R, Hernández-Chirlaque C, Sánchez de Medina F, Martínez-Augustin O. miR-146a regulates the crosstalk between intestinal epithelial cells, microbial components and inflammatory stimuli. Sci Rep 2018; 8:17350. [PMID: 30478292 PMCID: PMC6255912 DOI: 10.1038/s41598-018-35338-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 08/14/2018] [Indexed: 12/15/2022] Open
Abstract
Regulation of miR-146a abundance and its role in intestinal inflammation and particularly in intestinal epithelial cells (IECs) has been poorly studied. Here we study the relationship between bacterial antigens and inflammatory stimuli, and miR-146a expression using IEC lines and models of colitis (trinitrobenzenesulfonic acid (TNBS), dextran sulfate sodium (DSS) and the CD4 + CD62L + T cell transfer model). Specific bacterial antigens and cytokines (LPS, flagelin and IL-1β/TNF) stimulate miR-146a expression, while peptidoglycan, muramyldipeptide and CpG DNA have no effect. Overexpression of miR-146a by LPS depends on the activation of the TLR4/MyD88/NF-kB and Akt pathways. Accordingly, the induction of miR-146a is lower in TLR4, but not in TLR2 knock out mice in both basal and colitic conditions. miR-146a overexpression in IECs induces immune tolerance, inhibiting cytokine production (MCP-1 and GROα/IL-8) in response to LPS (IEC18) or IL-1β (Caco-2). Intestinal inflammation induced by chemical damage to the epithelium (DSS and TNBS models) induces miR-146a, but no effect is observed in the lymphocyte transfer model. Finally, we found that miR-146a expression is upregulated in purified IECs from villi vs. crypts. Our results indicate that miR-146a is a key molecule in the interaction among IECs, inflammatory stimuli and the microbiota.
Collapse
Grants
- BFU2014-57736-P, SAF2011-22922 Ministerio de Economía y Competitividad (Ministry of Economy and Competitiveness)
- AGL2014-5883-R, SAF-2011-22812 Ministerio de Economía y Competitividad (Ministry of Economy and Competitiveness)
- SAF2017-88457-R, BFU2014-57736-P, SAF2011-22922 Ministerio de Economía y Competitividad (Ministry of Economy and Competitiveness)
- CTS-245, CTS-6736 Consejería de Economía, Innovación, Ciencia y Empleo, Junta de Andalucía (Ministry of Economy, Innovation, Science and Employment, Government of Andalucia)
- CTS-235, CTS-6736 Consejería de Economía, Innovación, Ciencia y Empleo, Junta de Andalucía (Ministry of Economy, Innovation, Science and Employment, Government of Andalucia)
- CTS-245, CTS-6736 Consejería de Economía, Innovación, Ciencia y Empleo, Junta de Andalucía (Ministry of Economy, Innovation, Science and Employment, Government of Andalucia)
- Ministerio de Econom&#x00ED;a y Competitividad (Ministry of Economy and Competitiveness)
- Consejer&#x00ED;a de Econom&#x00ED;a, Innovaci&#x00F3;n, Ciencia y Empleo, Junta de Andaluc&#x00ED;a (Ministry of Economy, Innovation, Science and Employment, Government of Andalucia)
Collapse
Affiliation(s)
- Andrea Anzola
- Department of Pharmacology, CIBERehd, School of Pharmacy, Instituto de Investigación Biosanitaria ibs.GRANADA, University of Granada, Granada, Spain
| | - Raquel González
- Department of Pharmacology, CIBERehd, School of Pharmacy, Instituto de Investigación Biosanitaria ibs.GRANADA, University of Granada, Granada, Spain
| | - Reyes Gámez-Belmonte
- Department of Pharmacology, CIBERehd, School of Pharmacy, Instituto de Investigación Biosanitaria ibs.GRANADA, University of Granada, Granada, Spain
| | - Borja Ocón
- Department of Pharmacology, CIBERehd, School of Pharmacy, Instituto de Investigación Biosanitaria ibs.GRANADA, University of Granada, Granada, Spain
| | - Carlos J Aranda
- Department of Biochemistry and Molecular Biology II, CIBERehd, School of Pharmacy, Instituto de Investigación Biosanitaria ibs.GRANADA, University of Granada, Granada, Spain
| | - Patricia Martínez-Moya
- Department of Biochemistry and Molecular Biology II, CIBERehd, School of Pharmacy, Instituto de Investigación Biosanitaria ibs.GRANADA, University of Granada, Granada, Spain
| | - Rocío López-Posadas
- Department of Pharmacology, CIBERehd, School of Pharmacy, Instituto de Investigación Biosanitaria ibs.GRANADA, University of Granada, Granada, Spain
| | - Cristina Hernández-Chirlaque
- Department of Biochemistry and Molecular Biology II, CIBERehd, School of Pharmacy, Instituto de Investigación Biosanitaria ibs.GRANADA, University of Granada, Granada, Spain
| | - Fermín Sánchez de Medina
- Department of Pharmacology, CIBERehd, School of Pharmacy, Instituto de Investigación Biosanitaria ibs.GRANADA, University of Granada, Granada, Spain.
| | - Olga Martínez-Augustin
- Department of Biochemistry and Molecular Biology II, CIBERehd, School of Pharmacy, Instituto de Investigación Biosanitaria ibs.GRANADA, University of Granada, Granada, Spain
| |
Collapse
|
59
|
Aranda CJ, Ocón B, Arredondo‐Amador M, Suárez MD, Zarzuelo A, Chazin WJ, Martínez‐Augustin O, Sánchez de Medina F. Calprotectin protects against experimental colonic inflammation in mice. Br J Pharmacol 2018; 175:3797-3812. [PMID: 30007036 PMCID: PMC6135788 DOI: 10.1111/bph.14449] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 03/23/2018] [Accepted: 03/24/2018] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND AND PURPOSE Calprotectin is a heterodimer composed of two myeloid-related proteins, S100A8 and S100A9, that is abundant in neutrophils and monocytes/macrophages. Faecal levels of calprotectin are used routinely to monitor inflammatory bowel disease activity. EXPERIMENTAL APPROACH We aimed to assess the role of calprotectin in intestinal inflammation, using the dextran sulfate sodium model of colitis in mice. Calprotectin was administered (50 or 100 μg·day-1 ) by the intrarectal or by i.p. injection (50 μg·day-1 only). The condition of the mice was characterized by morphological and biochemical methods. KEY RESULTS Intrarectal calprotectin protected significantly against colitis, as shown by lower levels of macroscopic and microscopic damage, colonic myeloperoxidase activity and decreased expression of TNFα and toll-like receptor 4. IL-17 production by spleen and mesenteric lymph node cells was reduced. Calprotectin had no effect on body weight loss or colonic thickening. There were no effects of calprotectin after i.p. injection. Calprotectin had virtually no effects in control, non-colitic mice. Calprotectin had almost no effect on the colonic microbiota but enhanced barrier function. Treatment of rat IEC18 intestinal epithelial cells in vitro with calprotectin induced output of the chemokines CXL1 and CCL2, involving the receptor for advanced glycation end products- and NFκB. CONCLUSION AND IMPLICATIONS Calprotectin exerted protective effects in experimental colitis when given by the intrarectal route, by actions that appear to involve effects on the epithelium.
Collapse
Affiliation(s)
- Carlos J Aranda
- Department of Biochemistry and Molecular Biology II, CIBERehd, School of Pharmacy, Instituto de Investigación Biosanitaria ibs.GRANADAUniversity of GranadaGranadaSpain
| | - Borja Ocón
- Department of Pharmacology, CIBERehd, School of Pharmacy, Instituto de Investigación Biosanitaria ibs.GRANADAUniversity of GranadaGranadaSpain
| | - María Arredondo‐Amador
- Department of Pharmacology, CIBERehd, School of Pharmacy, Instituto de Investigación Biosanitaria ibs.GRANADAUniversity of GranadaGranadaSpain
| | - María Dolores Suárez
- Department of Biochemistry and Molecular Biology II, CIBERehd, School of Pharmacy, Instituto de Investigación Biosanitaria ibs.GRANADAUniversity of GranadaGranadaSpain
| | - Antonio Zarzuelo
- Department of Pharmacology, CIBERehd, School of Pharmacy, Instituto de Investigación Biosanitaria ibs.GRANADAUniversity of GranadaGranadaSpain
| | - Walter J Chazin
- Department of Biochemistry and Chemistry, Center for Structural BiologyVanderbilt UniversityNashvilleTNUSA
| | - Olga Martínez‐Augustin
- Department of Biochemistry and Molecular Biology II, CIBERehd, School of Pharmacy, Instituto de Investigación Biosanitaria ibs.GRANADAUniversity of GranadaGranadaSpain
| | - Fermín Sánchez de Medina
- Department of Pharmacology, CIBERehd, School of Pharmacy, Instituto de Investigación Biosanitaria ibs.GRANADAUniversity of GranadaGranadaSpain
| |
Collapse
|
60
|
Wagenaar L, Bol‐Schoenmakers M, Giustarini G, Vonk MM, van Esch BC, Knippels LM, Garssen J, Smit JJ, Pieters RH. Dietary Supplementation with Nondigestible Oligosaccharides Reduces Allergic Symptoms and Supports Low Dose Oral Immunotherapy in a Peanut Allergy Mouse Model. Mol Nutr Food Res 2018; 62:e1800369. [PMID: 30102006 PMCID: PMC6766954 DOI: 10.1002/mnfr.201800369] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 06/15/2018] [Indexed: 12/31/2022]
Abstract
SCOPE A major downside of oral immunotherapy (OIT) for food allergy is the risk of severe side effects. Non-digestible short- and long-chain fructo-oligosaccharides (scFOS/lcFOS) reduce allergy development in murine models. Therefore, it is hypothesized that scFOS/lcFOS can also support the efficacy of OIT in a peanut allergy model. METHODS AND RESULTS After sensitization to peanut extract (PE) using cholera toxin, C3H/HeOuJ mice are fed a 1% scFOS/lcFOS or control diet and receive OIT (1.5 or 15 mg PE). Hereafter, mice are exposed to PE via different routes to determine the safety and efficacy of treatment in clinical outcomes, PE-specific antibody production, and numbers of various immune cells. scFOS/lcFOS increases short-chain fatty acid levels in the caecum and reduce the acute allergic skin response and drop in body temperature after PE exposure. Interestingly, 15 mg and 1.5 mg OIT with scFOS/lcFOS induce protection against anaphylaxis, whereas 1.5 mg OIT alone does not. OIT, with or without scFOS/lcFOS, induces PE-specific immunoglobulin (Ig) IgG and IgA levels and increases CD103+ dendritic cells in the mesenteric lymph nodes. CONCLUSIONS scFOS/lcFOS and scFOS/lcFOS combined with low dose OIT are able to protect against a peanut-allergic anaphylactic response.
Collapse
Affiliation(s)
- Laura Wagenaar
- Department of ImmunotoxicologyInstitute for Risk Assessment SciencesUtrecht UniversityYalelaan 104, NL‐3584 CMUtrechtThe Netherlands
| | - Marianne Bol‐Schoenmakers
- Department of ImmunotoxicologyInstitute for Risk Assessment SciencesUtrecht UniversityYalelaan 104, NL‐3584 CMUtrechtThe Netherlands
| | - Giulio Giustarini
- Department of ImmunotoxicologyInstitute for Risk Assessment SciencesUtrecht UniversityYalelaan 104, NL‐3584 CMUtrechtThe Netherlands
| | - Marlotte M. Vonk
- Division of PharmacologyUtrecht Institute for Pharmaceutical SciencesUtrecht UniversityUniversiteitsweg 99, NL‐3584 CGUtrechtThe Netherlands
- Immunology PlatformNutricia ResearchUppsalalaan 12, NL‐3584 CTUtrechtThe Netherlands
| | - Betty C.A.M. van Esch
- Division of PharmacologyUtrecht Institute for Pharmaceutical SciencesUtrecht UniversityUniversiteitsweg 99, NL‐3584 CGUtrechtThe Netherlands
- Immunology PlatformNutricia ResearchUppsalalaan 12, NL‐3584 CTUtrechtThe Netherlands
| | - Leon M.J. Knippels
- Division of PharmacologyUtrecht Institute for Pharmaceutical SciencesUtrecht UniversityUniversiteitsweg 99, NL‐3584 CGUtrechtThe Netherlands
- Immunology PlatformNutricia ResearchUppsalalaan 12, NL‐3584 CTUtrechtThe Netherlands
| | - Johan Garssen
- Division of PharmacologyUtrecht Institute for Pharmaceutical SciencesUtrecht UniversityUniversiteitsweg 99, NL‐3584 CGUtrechtThe Netherlands
- Immunology PlatformNutricia ResearchUppsalalaan 12, NL‐3584 CTUtrechtThe Netherlands
| | - Joost J. Smit
- Department of ImmunotoxicologyInstitute for Risk Assessment SciencesUtrecht UniversityYalelaan 104, NL‐3584 CMUtrechtThe Netherlands
| | - Raymond H.H. Pieters
- Department of ImmunotoxicologyInstitute for Risk Assessment SciencesUtrecht UniversityYalelaan 104, NL‐3584 CMUtrechtThe Netherlands
| |
Collapse
|
61
|
Dai Z, Lyu W, Xiang X, Tang Y, Hu B, Ou S, Zeng X. Immunomodulatory Effects of Enzymatic-Synthesized α-Galactooligosaccharides and Evaluation of the Structure-Activity Relationship. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:9070-9079. [PMID: 30086236 DOI: 10.1021/acs.jafc.8b01939] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
In this study, α-galactooligosaccharides (α-GOSs) were synthesized using galactose as the substrate and α-galactosidase from Aspergillus niger as the catalyst. In the reaction, synthesized products of U1, U2, U3, and U4 were detected by high-performance liquid chromatography. By mass spectrometry, nuclear magnetic resonance, and 1-phenyl-3-methyl-5-pyrazolone derivatization, U1 was the mixture of disaccharides of α-d-Gal p-(1→1)-α-d-Gal, α-d-Gal p-(1→2)-α-d-Gal, α-d-Gal p-(1→3)-α-d-Gal, α-d-Gal p-(1→4)-α-d-Gal, U2 was identified to be α-d-Gal p-(1→6)-α-d-Gal, U3 was the mixture of galactotrisaccharides linked by one α-(1→6)-glycosidic linkage and one other α-glycosidic linkage, and U4 was identified as α-d-Gal p-(1→6)-α-d-Gal p-(1→6)-α-d-Gal. Afterward, the synthesized α-GOSs (U1, U2, U3, U4, and their mixture) as well as α-GOSs (manninotriose, stachyose, ciceritol, and verbascose) obtained from natural materials were used as subjects to evaluate their immunomodulatory effects in vitro by culturing mouse macrophage RAW264.7 cells. The results showed that α-GOS with a higher degree of polymerization had better immunomodulatory activity, while to a certain extent, α-GOS linked with α-(1→6)-galactosidic linkage showed a better immunomodulatory effect.
Collapse
Affiliation(s)
- Zhuqing Dai
- College of Food Science and Technology , Nanjing Agricultural University , Nanjing , Jiangsu 210095 , People's Republic of China
- Institute of Farm Product Processing , Jiangsu Academy of Agricultural Sciences , Nanjing , Jiangsu 210014 , People's Republic of China
| | - Wanyong Lyu
- Nutrition and Food Branch of China Association of Gerontology and Geriatrics , Beijing 100050 , People's Republic of China
| | - Xiaoli Xiang
- College of Food Science and Technology , Nanjing Agricultural University , Nanjing , Jiangsu 210095 , People's Republic of China
| | - Yuhong Tang
- College of Food Science and Technology , Nanjing Agricultural University , Nanjing , Jiangsu 210095 , People's Republic of China
| | - Bing Hu
- College of Food Science and Technology , Nanjing Agricultural University , Nanjing , Jiangsu 210095 , People's Republic of China
| | - Shiyi Ou
- Department of Food Science and Engineering , Jinan University , Guangzhou , Guangdong 510632 , People's Republic of China
| | - Xiaoxiong Zeng
- College of Food Science and Technology , Nanjing Agricultural University , Nanjing , Jiangsu 210095 , People's Republic of China
| |
Collapse
|
62
|
Cian RE, Hernández-Chirlaque C, Gámez-Belmonte R, Drago SR, Sánchez de Medina F, Martínez-Augustin O. Green Alga Ulva spp. Hydrolysates and Their Peptide Fractions Regulate Cytokine Production in Splenic Macrophages and Lymphocytes Involving the TLR4-NFκB/MAPK Pathways. Mar Drugs 2018; 16:E235. [PMID: 29997311 PMCID: PMC6071126 DOI: 10.3390/md16070235] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 06/27/2018] [Accepted: 07/10/2018] [Indexed: 01/01/2023] Open
Abstract
Hydrolysates of food protein sources have immunomodulatory effects, which are of interest for use as functional foods. In this study, we have characterized the immune regulatory effect on rat splenocytes, macrophages and T lymphocytes of Ulva spp. hydrolysates and their peptide fractions with or without in vitro gastrointestinal digestion and/or ultrafiltration. IL-10 was induced in almost all conditions and cell types obtained from wild type animals. The induction was in general increased by ultrafiltration and in vitro gastrointestinal digestion. TNF was also induced in basal conditions. In turn, TNF and IFN-γ production was attenuated by the hydrolysate products in lipopolysaccharide or concanavalin A immune stimulated cells. Inhibitors for the activation of NFκB, MAPK p38 and JNK inhibited IL-10 induction in rat splenocytes. The response was dramatically attenuated in TLR4-/- cells, and only modestly in TLR2-/- cells. Food peptides from Ulva spp. genus exert anti-inflammatory effects in immune cells mediated by TLR4 and NFκB. Similarity with the immunomodulatory profile of protein hydrolysates from other sources suggests a common mechanism.
Collapse
Affiliation(s)
- Raúl E Cian
- Instituto de Tecnología de Alimentos, CONICET, FIQ-UNL, 1 de Mayo 3250, 3000 Santa Fe, Argentina.
| | - Cristina Hernández-Chirlaque
- Department of Biochemistry and Molecular Biology II, CIBERehd, School of Pharmacy, Instituto de Investigación Biosanitaria ibs.GRANADA, University of Granada, 18071 Granada, Spain.
| | - Reyes Gámez-Belmonte
- Department of Pharmacology, CIBERehd, School of Pharmacy, Instituto de Investigación Biosanitaria ibs.GRANADA, University of Granada, 18071 Granada, Spain.
| | - Silvina R Drago
- Instituto de Tecnología de Alimentos, CONICET, FIQ-UNL, 1 de Mayo 3250, 3000 Santa Fe, Argentina.
| | - Fermín Sánchez de Medina
- Department of Pharmacology, CIBERehd, School of Pharmacy, Instituto de Investigación Biosanitaria ibs.GRANADA, University of Granada, 18071 Granada, Spain.
| | - Olga Martínez-Augustin
- Department of Biochemistry and Molecular Biology II, CIBERehd, School of Pharmacy, Instituto de Investigación Biosanitaria ibs.GRANADA, University of Granada, 18071 Granada, Spain.
| |
Collapse
|
63
|
Perdijk O, van Neerven RJJ, van den Brink E, Savelkoul HFJ, Brugman S. The oligosaccharides 6'-sialyllactose, 2'-fucosyllactose or galactooligosaccharides do not directly modulate human dendritic cell differentiation or maturation. PLoS One 2018; 13:e0200356. [PMID: 29990329 PMCID: PMC6039038 DOI: 10.1371/journal.pone.0200356] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 06/25/2018] [Indexed: 01/25/2023] Open
Abstract
Breast milk plays an important role in immune development in early life and protects against diseases later in life. A wide range of the beneficial effects of breast milk are attributed to human milk oligosaccharides (HMOs) as well as components such as vitamin D3 (VitD3) or TGFβ. One mechanism by which HMOs might contribute to immune homeostasis and protection against disease is the induction of a local tolerogenic milieu. In this study we investigated the effect of the HMOs 6’-sialyllactose (6’SL) and 2’-fucosyllactose (2’FL) as well as prebiotic galactooligosaccharides (GOS) on DC differentiation and maturation. Isolated CD14+ monocytes were cultured for six days in the presence of GM-CSF and IL-4 with or without 6’SL, 2’FL, GOS, VitD3 or TGFβ. Additionally, immature VitD3DC, TGFβDC and moDC were used as different DC types to investigate the effect of 6’SL, 2’FL and GOS on DC maturation. Surface marker expression and cytokine production was measured by flow cytometry and cytometric bead array, respectively. Unlike TGFβ and vitD3, the oligosaccharides 6’SL, 2’FL and GOS did not influence DC differentiation. Next, we studied the effect of 6’SL, 2’FL and GOS on maturation of moDC, VitD3DC and TGFβDC that showed different profiles of HMO-binding receptors. 6’SL, 2’FL and GOS did not modulate LPS-induced maturation, even though their putative receptors were present on the different DCs types. Thus, whereas VitD3 and TGFβ halt DC differentiation, which results in phenotypically distinct tolerogenic DCs, 6’SL, 2’FL and GOS do not alter DC differentiation or maturation of in vitro differentiated DC types.
Collapse
Affiliation(s)
- Olaf Perdijk
- Cell Biology and Immunology group, Wageningen University, Wageningen, the Netherlands
| | - R. J. Joost van Neerven
- Cell Biology and Immunology group, Wageningen University, Wageningen, the Netherlands
- FrieslandCampina, Amersfoort, the Netherlands
| | - Erik van den Brink
- Cell Biology and Immunology group, Wageningen University, Wageningen, the Netherlands
| | - Huub F. J. Savelkoul
- Cell Biology and Immunology group, Wageningen University, Wageningen, the Netherlands
| | - Sylvia Brugman
- Cell Biology and Immunology group, Wageningen University, Wageningen, the Netherlands
- * E-mail:
| |
Collapse
|
64
|
Wan Y, Fu Y, Wang F, Sinclair AJ, Li D. Protective Effects of a Lipid Extract from Hard-Shelled Mussel ( Mytilus coruscus) on Intestinal Integrity after Lipopolysaccharide Challenge in Mice. Nutrients 2018; 10:nu10070860. [PMID: 29970837 PMCID: PMC6073703 DOI: 10.3390/nu10070860] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Revised: 06/27/2018] [Accepted: 07/01/2018] [Indexed: 01/09/2023] Open
Abstract
This study investigated the protective effects of a lipid extract from hard-shelled mussel (HMLE) on intestinal integrity and the underlying mechanisms after a lipopolysaccharide (LPS) challenge in mice by using a 3 × 2 factorial design. Mice received olive oil, fish oil, and HMLE (n = 12 per group) by using gastric gavage for six weeks, respectively. Then half the mice in each group was injected intraperitoneally with LPS and the other half with phosphate buffered saline. Four hours after injection, mice were sacrificed and samples were collected. n-3 PUFAs were significantly enriched in erythrocytes following fish oil and HMLE supplementation. Both fish oil and HMLE improved intestinal morphology by restoring the ileac villus height and barrier function, which is indicated by decreased colonic myeloperoxidase activity and increased diamine oxidase activity as well as enhanced mRNA expression of intestinal tight junction proteins known as occludin and claudin-1 when compared with olive oil. In addition, both fish oil and HMLE increased colon production and the expression of anti-inflammatory cytokine, IL-10, while they inhibited the abnormal production and expression of pro-inflammatory cytokines including TNF-α, IL-1β, and IL-6 relative to the olive oil. Lastly, in comparison with olive oil, both fish oil and HMLE downregulated the TLR-4 signaling pathway by reducing the expression of two key molecules in this pathway, which are called TLR-4 and MyD88. These results suggest that HMLE had a protective effect on intestinal integrity after the LPS challenge, which was equivalent to that of fish oil. This effect might be associated with the regulation of inflammatory mediators and the inhibition of the TLR-4 signaling pathway.
Collapse
Affiliation(s)
- Yi Wan
- Institution of Nutrition and Health, Qingdao University, Qingdao 266071, China.
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou 310058, China.
| | - Yuanqing Fu
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou 310058, China.
- Department of Maternal and Infant Nutrition Research, Beingmate Baby and Child Food Co., Ltd., Hangzhou 311106, China.
| | - Fenglei Wang
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou 310058, China.
| | - Andrew J Sinclair
- School of Medicine, Deakin University, Locked Bag, Geelong 20000, Australia.
| | - Duo Li
- Institution of Nutrition and Health, Qingdao University, Qingdao 266071, China.
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
65
|
Celiberto LS, Graef FA, Healey GR, Bosman ES, Jacobson K, Sly LM, Vallance BA. Inflammatory bowel disease and immunonutrition: novel therapeutic approaches through modulation of diet and the gut microbiome. Immunology 2018; 155:36-52. [PMID: 29693729 DOI: 10.1111/imm.12939] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 03/28/2018] [Accepted: 04/18/2018] [Indexed: 12/11/2022] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory condition of the gastrointestinal tract, thought to at least in part reflect an aberrant immune response to gut bacteria. IBD is increasing in incidence, particularly in populations that have recently immigrated to western countries. This suggests that environmental factors are involved in its pathogenesis. We hypothesize that the increase in IBD rates might reflect the consumption of an unhealthy Western diet, containing excess calories and lacking in key nutritional factors, such as fibre and vitamin D. Several recent studies have determined that dietary factors can dramatically influence the activation of immune cells and the mediators they release through a process called immunonutrition. Moreover, dietary changes can profoundly affect the balance of beneficial versus pathogenic bacteria in the gut. This microbial imbalance can alter levels of microbiota-derived metabolites that in turn can influence innate and adaptive intestinal immune responses. If the diet-gut microbiome disease axis does indeed underpin much of the 'western' influence on the onset and progression of IBD, then tremendous opportunity exists for therapeutic changes in lifestyle, to modulate the gut microbiome and to correct immune imbalances in individuals with IBD. This review highlights four such therapeutic strategies - probiotics, prebiotics, vitamin D and caloric restriction - that have the potential to improve and add to current IBD treatment regimens.
Collapse
Affiliation(s)
- Larissa S Celiberto
- Department of Paediatrics, BC Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Franziska A Graef
- Department of Paediatrics, BC Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Genelle R Healey
- Department of Paediatrics, BC Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Else S Bosman
- Department of Paediatrics, BC Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Kevan Jacobson
- Department of Paediatrics, BC Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Laura M Sly
- Department of Paediatrics, BC Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Bruce A Vallance
- Department of Paediatrics, BC Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
66
|
Kopp TI, Vogel U, Tjonneland A, Andersen V. Meat and fiber intake and interaction with pattern recognition receptors (TLR1, TLR2, TLR4, and TLR10) in relation to colorectal cancer in a Danish prospective, case-cohort study. Am J Clin Nutr 2018; 107:465-479. [PMID: 29566186 DOI: 10.1093/ajcn/nqx011] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 10/20/2017] [Indexed: 12/13/2022] Open
Abstract
Background Meat and dietary fiber are associated with increased and decreased risk of colorectal cancer (CRC), respectively. Toll-like receptors (TLRs) regulate the intestinal immune response in a complex interplay between the mucosal epithelium and the microbiota and may therefore be important modulators of diet-induced CRC together with other inflammatory mediators. Objective Our aim was to investigate the association between functional TLR polymorphisms and risk of CRC and the interaction with dietary factors. Additionally, interactions with previously studied polymorphisms in IL10, IL1B, PTGS2, and NFKB1 were assessed in order to examine possible biological pathways in meat-induced CRC. Design A nested case-cohort study of 897 CRC cases and 1689 randomly selected participants from the Danish prospective "Diet, Cancer and Health" study encompassing 57,053 persons was performed using Cox proportional hazard models and the likelihood ratio test. Results We found associations between polymorphisms in TLR2 (P = 0.018) and TLR4 (P = 0.044) and risk of CRC per se, interactions between intake of red and processed meat (10 g/d) and polymorphisms in TLR1 (P-interaction = 0.032) and TLR10 (P-interaction = 0.026 and 0.036), and intake of cereals (50 g/d) and TLR4 (P-interaction = 0.044) in relation to risk of CRC. Intake of red and processed meat also interacted with combinations of polymorphisms in TLR1 and TLR10 and polymorphisms in NFKB1, IL10, IL1B, and PTGS2 (P-interaction; TLR1/rs4833095 × PTGS2/rs20417 = 0.021, TLR10/rs11096955 × IL10/rs3024505 = 0.047, TLR10/rs11096955 × PTGS2/rs20417 = 0.017, TLR10/rs4129009 × NFKB1/rs28362491 = 0.027, TLR10/rs4129009 × IL1B/rs4848306 = 0.020, TLR10/rs4129009 × IL1B/rs1143623 = 0.021, TLR10/rs4129009 × PTGS2/rs20417 = 0.027), whereas intake of dietary fiber (10 g/d) interacted with combinations of polymorphisms in TLR4, IL10, and PTGS2 (P-interaction; TLR4/rs1554973 × IL10/rs3024505 = 0.0012, TLR4/rs1554973 × PTGS2/rs20417 = 0.0041, TLR4/rs1554973 × PTGS2/rs5275 = 0.0064). Conclusions Our study suggests that meat intake may activate TLRs at the epithelial surface, leading to CRC via inflammation by nuclear transcription factor-κB-initiated transcription of inflammatory genes, whereas intake of fiber may protect against CRC via TLR4-mediated secretion of interleukin-10 and cyclooxygenase-2. Our results should be replicated in other prospective cohorts with well-characterized participants. The trial was registered at www.clinicaltrials.gov as NCT03250637.
Collapse
Affiliation(s)
- Tine Iskov Kopp
- Research Centre for Prevention and Health, Rigshospitalet-Glostrup, Glostrup, Denmark.,Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Ulla Vogel
- National Research Centre for the Working Environment, Copenhagen, Denmark
| | | | - Vibeke Andersen
- Focused Research Unit for Molecular Diagnostic and Clinical Research, Laboratory Center, Hospital of Southern Jutland, Aabenraa, Denmark.,Institute of Regional Health Research-Center Sønderjylland.,Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
67
|
Zehra S, Khambati I, Vierhout M, Mian MF, Buck R, Forsythe P. Human Milk Oligosaccharides Attenuate Antigen-Antibody Complex Induced Chemokine Release from Human Intestinal Epithelial Cell Lines. J Food Sci 2018; 83:499-508. [PMID: 29377120 DOI: 10.1111/1750-3841.14039] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 10/24/2017] [Accepted: 12/11/2017] [Indexed: 12/17/2022]
Abstract
There has been increased interest in the use of dietary ingredients, including prebiotics such as human-milk oligosaccharides (HMOs), as therapeutic strategies for food allergy. Understanding the mechanisms underlying the beneficial effects of HMOs is important to realizing their therapeutic potential. Here we demonstrate that the HMO, 6'-sialyllactose (6'SL) inhibited chemokine (IL-8 and CCL20) release from T-84 and HT-29 cells stimulated with antigen-antibody complex, TNFα or PGE2 ; an effect that was PPARγ dependent and associated with decreased activity of the transcription factors AP-1 and NFκB. In contrast, 2'-fucosyllactose (2'FL) selectively inhibited CCL20 release in response to antigen antibody complex in a PPARγ independent manner. This study reinforces the concept that structurally different oligosaccharides have distinct biological activities and identifies, for the first time, that the HMOs, 6'SL, and 2'FL, modulate human epithelial cell responses related to allergic disease. These findings encourage further investigation of the therapeutic potential of specific HMOs in food allergy. PRACTICAL APPLICATION This study provides evidence for direct effects of HMOs in addition to their prebiotic role and demonstrates, for the first time, modulation of Ag-IgE complex activation of human epithelial cells that may have important implications for food-allergy. The study also reinforces the concept that structurally different oligosaccharides have distinct biological activities. In determining the composition of infant formula, addition of oligosaccharides with specific structures may provide direct modulation of immune responses and potentially attenuate symptoms or development of food allergy.
Collapse
Affiliation(s)
- Sehrish Zehra
- Dept. of Medicine, Pathology and Molecular Medicine, McMaster Univ., Hamilton, Ont., Canada
| | - Ibrahim Khambati
- Dept. of Medicine, Pathology and Molecular Medicine, McMaster Univ., Hamilton, Ont., Canada
| | - Megan Vierhout
- Dept. of Medicine, Pathology and Molecular Medicine, McMaster Univ., Hamilton, Ont., Canada
| | - M Firoz Mian
- Dept. of Pathology and Molecular Medicine, McMaster Univ., Hamilton, Ont., Canada
| | - Rachael Buck
- Abbott Nutrition: a Division of Abbott Laboratories, Columbus, Ohio, U.S.A
| | - Paul Forsythe
- Dept. of Medicine, Pathology and Molecular Medicine, McMaster Univ., Hamilton, Ont., Canada.,The Firestone Inst. for Respiratory Research, McMaster Univ., Hamilton, Ont., Canada
| |
Collapse
|
68
|
Triantis V, Bode L, van Neerven RJJ. Immunological Effects of Human Milk Oligosaccharides. Front Pediatr 2018; 6:190. [PMID: 30013961 PMCID: PMC6036705 DOI: 10.3389/fped.2018.00190] [Citation(s) in RCA: 204] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 06/08/2018] [Indexed: 12/15/2022] Open
Abstract
Human milk oligosaccharides (HMOs) comprise a group of structurally complex, unconjugated glycans that are highly abundant in human milk. HMOs are minimally digested in the gastrointestinal tract and reach the colon intact, where they shape the microbiota. A small fraction of HMOs is absorbed, reaches the systemic circulation, and is excreted in urine. HMOs can bind to cell surface receptors expressed on epithelial cells and cells of the immune system and thus modulate neonatal immunity in the infant gut, and possibly also sites throughout the body. In addition, they have been shown to act as soluble decoy receptors to block the attachment of various microbial pathogens to cells. This review summarizes the current knowledge of the effects HMOs can have on infections, allergies, auto-immune diseases and inflammation, and will focus on the role of HMOs in altering immune responses through binding to immune-related receptors.
Collapse
Affiliation(s)
| | - Lars Bode
- Department of Pediatrics, University of California, San Diego, San Diego, CA, United States
| | - R J Joost van Neerven
- FrieslandCampina, Amersfoort, Netherlands.,Wageningen University and Research, Cell Biology and Immunology, Wageningen, Netherlands
| |
Collapse
|
69
|
Perdijk O, van Neerven RJJ, Meijer B, Savelkoul HFJ, Brugman S. Induction of human tolerogenic dendritic cells by 3′-sialyllactose via TLR4 is explained by LPS contamination. Glycobiology 2017; 28:126-130. [PMID: 29281012 PMCID: PMC5993091 DOI: 10.1093/glycob/cwx106] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Indexed: 01/05/2023] Open
Abstract
The human milk oligosaccharide 3′-sialyllactose (3′SL) has previously been shown to activate murine dendritic cells (DC) in a Toll-like receptor (TLR) 4-mediated manner ex vivo. In this study we aimed to investigate whether 3′SL has similar immunomodulatory properties on human DC. 3′SL was shown to induce NF-κB activation via human TLR4. However, LPS was detected in the commercially obtained 3′SL from different suppliers. After the removal of LPS from 3′SL, we studied its ability to modify DC differentiation in vitro. In contrast to LPS and 3′SL, LPS-free 3′SL did not induce functional and phenotypical changes on immature DC (iDC). iDC that were differentiated in the presence of LPS or 3′SL showed a semi-mature phenotype (i.e., fewer CD83+CD86+ DC), produced IL-10 and abrogated IL-12p70 and tumor necrosis factor-alpha levels upon stimulation with several TLR ligands. Differentiation into these tolerogenic DC was completely abrogated by LPS removal from 3′SL. In contrast to previous reports in mice, we found that LPS-free 3′SL does not activate NF-κB via human TLR4. In conclusion, removing LPS from (oligo)saccharide preparations is necessary to study their potential immunomodulatory function.
Collapse
Affiliation(s)
- Olaf Perdijk
- Cell Biology and Immunology group, Wageningen University & Research, Zodiac building 122, de Elst 1, 6708 WD, Wageningen, The Netherlands
| | - R J Joost van Neerven
- Cell Biology and Immunology group, Wageningen University & Research, Zodiac building 122, de Elst 1, 6708 WD, Wageningen, The Netherlands
- FrieslandCampina, Stationsplein 4, 3818 LE, Amersfoort, The Netherlands
| | - Ben Meijer
- Cell Biology and Immunology group, Wageningen University & Research, Zodiac building 122, de Elst 1, 6708 WD, Wageningen, The Netherlands
| | - Huub F J Savelkoul
- Cell Biology and Immunology group, Wageningen University & Research, Zodiac building 122, de Elst 1, 6708 WD, Wageningen, The Netherlands
| | - Sylvia Brugman
- Cell Biology and Immunology group, Wageningen University & Research, Zodiac building 122, de Elst 1, 6708 WD, Wageningen, The Netherlands
| |
Collapse
|
70
|
Chen Q, Ren Y, Lu J, Bartlett M, Chen L, Zhang Y, Guo X, Liu C. A Novel Prebiotic Blend Product Prevents Irritable Bowel Syndrome in Mice by Improving Gut Microbiota and Modulating Immune Response. Nutrients 2017; 9:nu9121341. [PMID: 29232851 PMCID: PMC5748791 DOI: 10.3390/nu9121341] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 11/24/2017] [Accepted: 12/07/2017] [Indexed: 02/06/2023] Open
Abstract
Irritable bowel syndrome (IBS) is the most common functional gastrointestinal disorder yet it still lacks effective prevention therapies. The aim of this study is to determine whether a novel prebiotic blend (PB) composed of fructo-oligosaccharide (FOS), galactooligosaccharide (GOS), inulin and anthocyanins could be effective in preventing the development of IBS. We explored the possible mechanisms both in animal and in cells. Post-infectious IBS models in C57BL/6 mice were established and were pretreated with the PB, PB and probiotic strains 8 weeks in advance of infection. Eight weeks after infection, intestinal tissues were collected for assessing histomorphology, visceral sensitivity, barrier function, pro-inflammatory cytokines expression and proteomics analysis. Fecal samples were also collected for microbiota analysis. The pro-inflammatory cytokines expression in Caco-2 cells were evaluated after co-incubation with PB and Salmonella typhimurium 14028. The results showed that PB significantly decreased the pro-inflammatory cytokines both in infected Caco-2 cells and PI-IBS models. The loss of body weight, decreased expression of tight junction protein Occludin (OCLN), and changes of the microbiota composition induced by infections could be greatly improved by PB intervention (p < 0.05). The proteomics analysis revealed that this function was associated with Peroxisome proliferator-activated receptor (PPAR)γ pathway.
Collapse
Affiliation(s)
- Qian Chen
- Department of Microbiology and Immunology, Institutes of Medical Science, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Yiping Ren
- Center for Anti-Aging Research, Nu Skin Enterprises, Shanghai 201401, China.
| | - Jihong Lu
- Center for Anti-Aging Research, Nu Skin Enterprises, Shanghai 201401, China.
| | - Mark Bartlett
- Nu Skin Enterprises Anti-Aging Research Center, Provo, UT 84601, USA.
| | - Lei Chen
- Department of Microbiology and Immunology, Institutes of Medical Science, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Yan Zhang
- Department of Microbiology and Immunology, Institutes of Medical Science, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Xiaokui Guo
- Department of Microbiology and Immunology, Institutes of Medical Science, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Chang Liu
- Department of Microbiology and Immunology, Institutes of Medical Science, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| |
Collapse
|
71
|
Zhang S, Yang J, Henning SM, Lee R, Hsu M, Grojean E, Pisegna R, Ly A, Heber D, Li Z. Dietary pomegranate extract and inulin affect gut microbiome differentially in mice fed an obesogenic diet. Anaerobe 2017; 48:184-193. [DOI: 10.1016/j.anaerobe.2017.08.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 08/30/2017] [Accepted: 08/31/2017] [Indexed: 12/27/2022]
|
72
|
Macrophages treated with non-digestible polysaccharides reveal a transcriptionally unique phenotype. J Funct Foods 2017. [DOI: 10.1016/j.jff.2017.07.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|
73
|
Akbari P, Fink-Gremmels J, Willems RHAM, Difilippo E, Schols HA, Schoterman MHC, Garssen J, Braber S. Characterizing microbiota-independent effects of oligosaccharides on intestinal epithelial cells: insight into the role of structure and size : Structure-activity relationships of non-digestible oligosaccharides. Eur J Nutr 2017; 56:1919-1930. [PMID: 27295033 PMCID: PMC5534205 DOI: 10.1007/s00394-016-1234-9] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 05/20/2016] [Indexed: 01/03/2023]
Abstract
PURPOSE The direct effects of galacto-oligosaccharides (GOS), including Vivinal® GOS syrup (VGOS) and purified Vivinal® GOS (PGOS), on the epithelial integrity and corresponding interleukin-8 (IL-8/CXCL8) release were examined in a Caco-2 cell model for intestinal barrier dysfunction. To investigate structure-activity relationships, the effects of individual DP fractions of VGOS were evaluated. Moreover, the obtained results with GOS were compared with Caco-2 monolayers incubated with fructo-oligosaccharides (FOS) and inulin. METHODS Caco-2 monolayers were pretreated (24 h) with or without specific oligosaccharides or DP fractions of VGOS (DP2 to DP6) before being exposed for 12 or 24 h to the fungal toxin deoxynivalenol (DON). Transepithelial electrical resistance and lucifer yellow permeability were measured to investigate barrier integrity. A calcium switch assay was used to study the reassembly of tight junction proteins. Release of CXCL8, a typical marker for inflammation, was quantified by ELISA. RESULTS In comparison with PGOS, FOS and inulin, VGOS showed the most pronounced protective effect on the DON-induced impairment of the monolayer integrity, acceleration of the tight junction reassembly and the subsequent CXCL8 release. DP2 and DP3 in concentrations occurring in VGOS prevented the DON-induced epithelial barrier disruption, which could be related to their high prevalence in VGOS. However, no effects of the separate DP GOS fractions were observed on CXCL8 release. CONCLUSIONS This comparative study demonstrates the direct, microbiota-independent effects of oligosaccharides on the intestinal barrier function and shows the differences between individual galacto- and fructo-oligosaccharides. This microbiota-independent effect of oligosaccharides depends on the oligosaccharide structure, DP length and concentration.
Collapse
Affiliation(s)
- Peyman Akbari
- Division of Veterinary Pharmacology, Pharmacotherapy and Toxicology, Institute for Risk Assessment Sciences, Utrecht University, Yalelaan 104, 3584 CM, Utrecht, The Netherlands
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences Faculty of Science, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Johanna Fink-Gremmels
- Division of Veterinary Pharmacology, Pharmacotherapy and Toxicology, Institute for Risk Assessment Sciences, Utrecht University, Yalelaan 104, 3584 CM, Utrecht, The Netherlands
| | - Rianne H A M Willems
- Laboratory of Food Chemistry, Wageningen University, 6708 WG, Wageningen, The Netherlands
| | - Elisabetta Difilippo
- Laboratory of Food Chemistry, Wageningen University, 6708 WG, Wageningen, The Netherlands
| | - Henk A Schols
- Laboratory of Food Chemistry, Wageningen University, 6708 WG, Wageningen, The Netherlands
| | | | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences Faculty of Science, Utrecht University, 3584 CG, Utrecht, The Netherlands
- Nutricia Research, 3584 CT, Utrecht, The Netherlands
| | - Saskia Braber
- Division of Veterinary Pharmacology, Pharmacotherapy and Toxicology, Institute for Risk Assessment Sciences, Utrecht University, Yalelaan 104, 3584 CM, Utrecht, The Netherlands.
| |
Collapse
|
74
|
|
75
|
Protein kinase C δ signaling is required for dietary prebiotic-induced strengthening of intestinal epithelial barrier function. Sci Rep 2017; 7:40820. [PMID: 28098206 PMCID: PMC5241689 DOI: 10.1038/srep40820] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 12/12/2016] [Indexed: 01/05/2023] Open
Abstract
Prebiotics are non-digestible oligosaccharides that promote the growth of beneficial gut microbes, but it is unclear whether they also have direct effects on the intestinal mucosal barrier. Here we demonstrate two commercial prebiotics, inulin and short-chain fructo-oligosaccharide (scFOS), when applied onto intestinal epithelia in the absence of microbes, directly promote barrier integrity to prevent pathogen-induced barrier disruptions. We further show that these effects involve the induction of select tight junction (TJ) proteins through a protein kinase C (PKC) δ-dependent mechanism. These results suggest that in the absence of microbiota, prebiotics can directly exert barrier protective effects by activating host cell signaling in the intestinal epithelium, which represents a novel alternative mechanism of action of prebiotics.
Collapse
|
76
|
Jiminez JA, Uwiera TC, Abbott DW, Uwiera RRE, Inglis GD. Impacts of resistant starch and wheat bran consumption on enteric inflammation in relation to colonic bacterial community structures and short-chain fatty acid concentrations in mice. Gut Pathog 2016; 8:67. [PMID: 28031748 PMCID: PMC5178079 DOI: 10.1186/s13099-016-0149-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 12/05/2016] [Indexed: 02/07/2023] Open
Abstract
Background
Identifying the connection among diet, the intestinal microbiome, and host health is currently an area of intensive research, but the potential of dietary fiber (DF) consumption to ameliorate intestinal inflammation has not been extensively studied. We examined the impacts of the DFs, wheat bran (WB) and resistant starch (RS) on host enteric health. A murine model of acute Th1/Th17 colitis (i.e. incited by Citrobacter rodentium) was used. Results Diets enriched with RS increased weight gain in mice inoculated with C. rodentium compared to mice consuming a conventional control (CN) diet. Short-chain fatty acid (SCFA) quantities in the cecum and distal colon were higher in mice consuming DFs, and these mice exhibited higher butyrate concentrations in the distal colon during inflammation. Histopathologic scores of inflammation in the proximal colon on day 14 post-inoculation (p.i.) (peak infection) and 21 p.i. (late infection) were lower in mice consuming DF-enriched diets compared to the CN diet. Consumption of WB reduced the expression of Th1/Th17 cytokines. As well, the expression of bacterial recognition and response genes such as Relmβ, RegIIIγ, and Tlr4 increased in mice consuming the RS-enriched diets. Furthermore, each diet generated a region-specific bacterial community, suggesting a link between selection for specific bacterial communities, SCFA concentrations, and inflammation in the murine colon. Conclusions Collectively, data indicated that the consumption of DF-rich diets ameliorates the effects of C. rodentium-induced enteritis by modifying the host microbiota to increase SCFA production, and bacterial recognition and response mechanisms to promote host health.
Electronic supplementary material The online version of this article (doi:10.1186/s13099-016-0149-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Janelle A Jiminez
- Agriculture and Agri-Food Canada, 5403-1st Avenue South, Lethbridge, AB T1J 4B1 Canada.,Department of Agricultural Food and Nutritional Science, University of Alberta, 410 Agriculture/Forestry Centre, Edmonton, AB T6G 2P5 Canada
| | - Trina C Uwiera
- Divisions of Pediatric Surgery, Department of Surgery, University of Alberta, 2C3.82 Walter C. Mackenzie Health Sciences Center, 8440-112th Street, Edmonton, AB T6G 2B7 Canada
| | - D Wade Abbott
- Agriculture and Agri-Food Canada, 5403-1st Avenue South, Lethbridge, AB T1J 4B1 Canada
| | - Richard R E Uwiera
- Department of Agricultural Food and Nutritional Science, University of Alberta, 410 Agriculture/Forestry Centre, Edmonton, AB T6G 2P5 Canada
| | - G Douglas Inglis
- Agriculture and Agri-Food Canada, 5403-1st Avenue South, Lethbridge, AB T1J 4B1 Canada
| |
Collapse
|
77
|
Johnson-Henry KC, Abrahamsson TR, Wu RY, Sherman PM. Probiotics, Prebiotics, and Synbiotics for the Prevention of Necrotizing Enterocolitis. Adv Nutr 2016; 7:928-37. [PMID: 27633108 PMCID: PMC5015037 DOI: 10.3945/an.116.012237] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Necrotizing enterocolitis (NEC) is a devastating intestinal disease in preterm infants characterized by barrier disruption, intestinal microbial dysbiosis, and persistent inflammation of the colon, which results in high mortality rates. Current strategies used to manage this disease are not sufficient, although the use of human breast milk reduces the risk of NEC. Mother's milk is regarded as a fundamental nutritional source for neonates, but pasteurization of donor breast milk affects the composition of bioactive compounds. Current research is evaluating the benefits and potential pitfalls of adding probiotics and prebiotics to pasteurized milk so as to improve the functionality of the milk and thereby reduce the burden of illness caused by NEC. Probiotics (live micro-organisms that confer health to the host) and prebiotics (nondigestible oligosaccharides that stimulate the growth of healthy bacteria) are functional foods known to mediate immune responses and modulate microbial populations in the gut. Clinical research shows strain- and compound-specific responses when probiotics or prebiotics are administered in conjunction with donor breast milk for the prevention of NEC. Despite ongoing controversy surrounding optimal treatment strategies, randomized controlled studies are now investigating the use of synbiotics to reduce the incidence and severity of NEC. Synbiotics, a combination of probiotics and prebiotics, have been proposed to enhance beneficial health effects in the intestinal tract more than either agent administered alone. This review considers the implications of using probiotic-, prebiotic-, and synbiotic-supplemented breast milk as a strategy to prevent NEC and issues that could be encountered with the preparations.
Collapse
Affiliation(s)
- Kathene C Johnson-Henry
- Cell Biology Program, Research Institute, Division of Gastroenterology, Hepatology, and Nutrition, Hospital for Sick Children
| | - Thomas R Abrahamsson
- Department of Clinical and Experimental Medicine, Division of Pediatrics, Linköping University, Linköping, Sweden
| | - Richard You Wu
- Cell Biology Program, Research Institute, Division of Gastroenterology, Hepatology, and Nutrition, Hospital for Sick Children;,Department of Laboratory Medicine and Pathobiology, Faculty of Medicine
| | - Philip M Sherman
- Cell Biology Program, Research Institute, Division of Gastroenterology, Hepatology, and Nutrition, Hospital for Sick Children; Department of Laboratory Medicine and Pathobiology, Faculty of Medicine; Department of Nutritional Sciences; and Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada; and
| |
Collapse
|
78
|
Immunoregulatory Effects Triggered by Lactic Acid Bacteria Exopolysaccharides: New Insights into Molecular Interactions with Host Cells. Microorganisms 2016; 4:microorganisms4030027. [PMID: 27681921 PMCID: PMC5039587 DOI: 10.3390/microorganisms4030027] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 06/10/2016] [Indexed: 01/02/2023] Open
Abstract
Researchers have demonstrated that lactic acid bacteria (LAB) with immunomodulatory capabilities (immunobiotics) exert their beneficial effects through several molecules, including cell wall, peptidoglycan, and exopolysaccharides (EPS), that are able to interact with specific host cell receptors. EPS from LAB show a wide heterogeneity in its composition, meaning that biological properties depend on the strain and. therefore, only a part of the mechanism of action has been elucidated for these molecules. In this review, we summarize the current knowledge of the health-promoting actions of EPS from LAB with special focus on their immunoregulatory actions. In addition, we describe our studies using porcine intestinal epithelial cells (PIE cells) as a model to evaluate the molecular interactions of EPS from two immunobiotic LAB strains and the host cells. Our studies showed that EPS from immunobiotic LAB have anti-inflammatory capacities in PIE cells since they are able to reduce the production of inflammatory cytokines in cells challenged with the Toll-like receptor (TLR)-4-agonist lipopolysaccharide. The effects of EPS were dependent on TLR2, TLR4, and negative regulators of TLR signaling. We also reported that the radioprotective 105 (RP105)/MD1 complex, a member of the TLR family, is partially involved in the immunoregulatory effects of the EPS from LAB. Our work described, for the first time, that LAB and their EPS reduce inflammation in intestinal epithelial cells in a RP105/MD1-dependent manner. A continuing challenge for the future is to reveal more effector-receptor relationships in immunobiotic-host interactions that contribute to the beneficial effects of these bacteria on mucosal immune homeostasis. A detailed molecular understanding should lead to a more rational use of immunobiotics in general, and their EPS in particular, as efficient prevention and therapies for specific immune-related disorders in humans and animals.
Collapse
|
79
|
Human milk oligosaccharides: The role in the fine-tuning of innate immune responses. Carbohydr Res 2016; 432:62-70. [PMID: 27448325 DOI: 10.1016/j.carres.2016.07.009] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 07/05/2016] [Accepted: 07/06/2016] [Indexed: 01/12/2023]
Abstract
In order to secure the health of newborns over the period of immune immaturity during the first months of life, a mother provides her offspring with passive protection: bioactive molecules transferred through the placenta and breast milk. It is well known that human milk contains immunoglobulins (Ig), immune cells and diverse cytokines, which affect newborn directly or indirectly and contribute to the maturation of the immune system. However, in addition to the above-stated molecules, human milk oligosaccharides (HMOs), a complex mixture of free indigestible carbohydrates with multiple functions, play exceptional roles in the functioning of the infants' immune system. These biological molecules have been studied over decades, however, interest in HMOs does not seem to have abated. Although biological activities of oligosaccharides from human milk have been explicitly reviewed, information regarding the role of HMOs in inflammation remains rather fragmented. The purpose of this review is to compile existing knowledge about the role of certain species of HMOs, including fucosylated, galactosylated and sialylated oligosaccharides, and their signaling pathways in immunity and inflammation. The advances in applying this information to the treatment of diseases in infants as well as adults were also reviewed here.
Collapse
|
80
|
Govers C, Tomassen MM, Rieder A, Ballance S, Knutsen SH, Mes JJ. Lipopolysaccharide quantification and alkali-based inactivation in polysaccharide preparations to enable in vitro immune modulatory studies. ACTA ACUST UNITED AC 2016. [DOI: 10.1016/j.bcdf.2016.09.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
81
|
van Esch BCAM, Abbring S, Diks MAP, Dingjan GM, Harthoorn LF, Vos AP, Garssen J. Post-sensitization administration of non-digestible oligosaccharides and Bifidobacterium breve M-16V reduces allergic symptoms in mice. Immun Inflamm Dis 2016; 4:155-165. [PMID: 27933160 PMCID: PMC4879462 DOI: 10.1002/iid3.101] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 01/26/2016] [Accepted: 01/27/2016] [Indexed: 12/16/2022] Open
Abstract
To support dietary management of severe cow's milk allergic infants, a synbiotic mixture of non-digestible oligosaccharides and Bifidobacterium breve M-16V (B. breve) was designed from source materials that are completely cow's milk-free. It was investigated whether this specific synbiotic concept can reduce an established food allergic response in a research model for hen's egg allergy. Mice were orally sensitized once a week for 5 weeks to ovalbumin (OVA) using cholera toxin (CT) as an adjuvant. Non-sensitized mice received CT in PBS only. Sensitized mice were fed a control diet or a diet enriched with short-chain- (scFOS) and long-chain fructo-oligosaccharides (lcFOS), B. breve or scFOSlcFOS + B. breve for 3 weeks starting after the last sensitization. Non-sensitized mice received the control diet. Anaphylactic shock symptoms, acute allergic skin responses and serum specific IgE, mMCP-1 and galectin-9 were measured upon OVA challenge. Activated Th2-, Th1-cells and regulatory T-cells were quantified in spleen and mesenteric lymph nodes (MLN) and cytokine profiles were analyzed. Short chain fatty acids (SCFA) were measured in ceacal samples. The acute allergic skin response was reduced in mice fed the scFOSlcFOS + B. breve diet compared to mice fed any of the other diets. A reduction in mast cell degranulation (mMCP-1) and anaphylactic shock symptoms was also observed in these mice. Unstimulated splenocyte cultures produced increased levels of IL10 and IFNg in mice fed the scFOSlcFOS + B. breve diet. Correspondingly, increased percentages of activated Th1 cells were observed in the spleen. Allergen-specific re-stimulation of splenocytes showed a decrease in IL5 production. In summary; post-sensitization administration of scFOSlcFOS + B. breve was effective in reducing allergic symptoms after allergen challenge. These effects coincided with changes in regulatory and effector T-cell subsets and increases in the SCFA propionic acid. These results suggest immune modulatory benefits of dietary intervention with a unique combination of scFOSlcFOS + B. breve in established food allergy. Whether these effects translate to human applications is subject for ongoing clinical studies.
Collapse
Affiliation(s)
- Betty C. A. M. van Esch
- Faculty of ScienceDivison of PharmacologyUtrecht Institute for Pharmaceutical SciencesUtrecht UniversityUtrechtThe Netherlands
- Nutricia ResearchUtrechtThe Netherlands
| | - Suzanne Abbring
- Faculty of ScienceDivison of PharmacologyUtrecht Institute for Pharmaceutical SciencesUtrecht UniversityUtrechtThe Netherlands
| | - Mara A. P. Diks
- Faculty of ScienceDivison of PharmacologyUtrecht Institute for Pharmaceutical SciencesUtrecht UniversityUtrechtThe Netherlands
| | - Gemma M. Dingjan
- Faculty of ScienceDivison of PharmacologyUtrecht Institute for Pharmaceutical SciencesUtrecht UniversityUtrechtThe Netherlands
| | | | - A. Paul Vos
- Faculty of ScienceDivison of PharmacologyUtrecht Institute for Pharmaceutical SciencesUtrecht UniversityUtrechtThe Netherlands
- Nutricia ResearchNutricia Advanced Medical NutritionUtrechtthe Netherlands
| | - Johan Garssen
- Faculty of ScienceDivison of PharmacologyUtrecht Institute for Pharmaceutical SciencesUtrecht UniversityUtrechtThe Netherlands
- Nutricia ResearchUtrechtThe Netherlands
| |
Collapse
|
82
|
The Gut Microbiota and Immune System Relationship in Human Graft-versus-Host Disease. Mediterr J Hematol Infect Dis 2016; 8:e2016025. [PMID: 27158438 PMCID: PMC4848019 DOI: 10.4084/mjhid.2016.025] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 04/10/2016] [Indexed: 02/07/2023] Open
Abstract
Gut microbiota has gained increasing interest in the pathogenesis of immune-related diseases. In this context, graft-versus-host disease is a condition characterized by an immune response which frequently complicates and limits the outcomes of hematopoietic stem cell transplantations. Past studies, carried mostly in animals, already supported a relationship between gut microbiota and graft-versus-host disease. However, the possible mechanisms underlying this connection remain elusory. Moreover, strategies to prevent graft-versus-host disease are of great interest as well as the potential role of gut microbiota modulation. We reviewed the role of gut microbiota in the development of immune system and its involvement in the graft-versus-host disease, focusing on data available on humans.
Collapse
|
83
|
Valcheva R, Dieleman LA. Prebiotics: Definition and protective mechanisms. Best Pract Res Clin Gastroenterol 2016; 30:27-37. [PMID: 27048894 DOI: 10.1016/j.bpg.2016.02.008] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 02/09/2016] [Accepted: 02/11/2016] [Indexed: 01/31/2023]
Abstract
The increase in chronic metabolic and immunologic disorders in the modern society is linked to major changes in the dietary patterns. These chronic conditions are associated with intestinal microbiota dysbiosis where important groups of carbohydrate fermenting, short-chain fatty acids-producing bacteria are reduced. Dietary prebiotics are defined as a selectively fermented ingredients that result in specific changes in the composition and/or activity of the gastrointestinal microbiota, thus conferring benefit(s) upon host health. Application of prebiotics may then restore the gut microbiota diversity and activity. Unlike the previously accepted prebiotics definition, where a limited number of bacterial species are involved in the prebiotic activity, new data from community-wide microbiome analysis demonstrated a broader affect of the prebiotics on the intestinal microbiota. These new findings require a revision of the current definition. In addition, prebiotics may exert immunomodulatory effects through microbiota-independent mechanisms that will require future investigations involving germ-free animal disease models.
Collapse
Affiliation(s)
- Rosica Valcheva
- Department of Medicine, Center of Excellence for Gastrointestinal Inflammation and Immunity Research, University of Alberta, AB, Canada.
| | - Levinus A Dieleman
- Department of Medicine, Center of Excellence for Gastrointestinal Inflammation and Immunity Research, University of Alberta, AB, Canada
| |
Collapse
|
84
|
|
85
|
Bernard H, Desseyn JL, Gottrand F, Stahl B, Bartke N, Husson MO. Pectin-Derived Acidic Oligosaccharides Improve the Outcome of Pseudomonas aeruginosa Lung Infection in C57BL/6 Mice. PLoS One 2015; 10:e0139686. [PMID: 26599638 PMCID: PMC4658080 DOI: 10.1371/journal.pone.0139686] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 09/16/2015] [Indexed: 02/07/2023] Open
Abstract
The administration of prebiotics as oligosaccharides (OS), by acting on intestinal microbiota, could modulate the immune and inflammatory response and represent a new strategy to improve the outcome of bacterial infection. The aim of this study was to determine whether pectin-derived acidic oligosaccharides (pAOS) could modulate the outcome of pulmonary P. aeruginosa (PA) infection in C57BL/6 mice, which develop a Th1 response to PA lung infection. Mice were randomized for 5 weeks to consume a control or a 5% pAOS diet and chronically infected by PA. Resistance to a second PA infection was also analyzed by reinfecting the surviving mice 2 weeks after the first infection. Compared with control mice, mice fed pAOS had reduced mortality (P<0.05). This improvement correlated with a better control of the inflammatory response with a lower neutrophil count on day 1 (P<0.05), a sustained neutrophil and macrophage recruitment on days 2 and 3 (P<0.01) a greater and sustained IL-10 release in lung (P<0.05) and a reduction of the Th1 response and M1 activation with a lower IFN-γ/IL-4 (P<0.01) and nos2/arg1 (P<0.05) ratios. These results coincided with a modulation of the intestinal microbiota as shown by an increased butyric acid concentration in feces (P<0.05). Moreover, pAOS decreased the bacterial load (P<0.01) in mice reinfected 2 weeks after the first infection, suggesting that pAOS could reduce pulmonary exacerbations. In conclusion, pAOS improved the outcome of PA infection in C57BL/6 mice by modulating the intestinal microbiota and the inflammatory and immune responses.
Collapse
Affiliation(s)
- Henry Bernard
- LIRIC UMR 995 Inserm; Université de Lille; CHRU de Lille, Faculté de Médecine, Place de Verdun, F-59045, Lille cedex, France
| | - Jean-Luc Desseyn
- LIRIC UMR 995 Inserm; Université de Lille; CHRU de Lille, Faculté de Médecine, Place de Verdun, F-59045, Lille cedex, France
| | - Frédéric Gottrand
- LIRIC UMR 995 Inserm; Université de Lille; CHRU de Lille, Faculté de Médecine, Place de Verdun, F-59045, Lille cedex, France
| | - Bernd Stahl
- Nutricia Research, Uppsalalaan 12, 3584 CT Utrecht, The Netherlands
| | - Nana Bartke
- Nutricia Research, Uppsalalaan 12, 3584 CT Utrecht, The Netherlands
| | - Marie-Odile Husson
- LIRIC UMR 995 Inserm; Université de Lille; CHRU de Lille, Faculté de Médecine, Place de Verdun, F-59045, Lille cedex, France
| |
Collapse
|
86
|
Varasteh S, Braber S, Akbari P, Garssen J, Fink-Gremmels J. Differences in Susceptibility to Heat Stress along the Chicken Intestine and the Protective Effects of Galacto-Oligosaccharides. PLoS One 2015; 10:e0138975. [PMID: 26402906 PMCID: PMC4581695 DOI: 10.1371/journal.pone.0138975] [Citation(s) in RCA: 173] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 09/07/2015] [Indexed: 01/13/2023] Open
Abstract
High ambient temperatures negatively affect the human well-being as well as animal welfare and production. The gastrointestinal tract is predominantly responsive to heat stress. The currently available information about the multifaceted response to heat stress within different parts of the intestine is limited, especially in avian species. Hence, this study aims to evaluate the heat stress-induced sequence of events in the intestines of chickens. Furthermore, the gut health-promoting effect of dietary galacto-oligosaccharides (GOS) was investigated in these heat stress-exposed chickens. Chickens were fed a control diet or diet supplemented with 1% or 2.5% GOS (6 days) prior to and during a temperature challenge for 5 days (38-39°C, 8h per day). The parameters measured in different parts of the intestines included the genes (qPCR) HSF1, HSF3, HSP70, HSP90, E-cadherin, claudin-1, claudin-5, ZO-1, occludin, TLR-2, TLR-4, IL-6, IL-8, HO-1, HIF-1α) and their associated proteins HSP70, HSP90 and pan-cadherin (western blots). In addition, IL-6 and IL-8 plasma concentrations were measured by ELISA. In the jejunum, HSF3, HSP70, HSP90, E-cadherin, claudin-5, ZO-1, TLR-4, IL-6 and IL-8 mRNA expression and HSP70 protein expression were increased after heat stress exposure and a more pronounced increase in gene expression was observed in ileum after heat stress exposure, and in addition HSF1, claudin-1 and HIF-1α mRNA levels were upregulated. Furthermore, the IL-8 plasma levels were decreased in chickens exposed to heat stress. Interestingly, the heat stress-related effects in the jejunum were prevented in chickens fed a GOS diet, while dietary GOS did not alter these effects in ileum. In conclusion, our results demonstrate the differences in susceptibility to heat stress along the intestine, where the most obvious modification in gene expression is observed in ileum, while dietary GOS only prevent the heat stress-related changes in jejunum.
Collapse
Affiliation(s)
- Soheil Varasteh
- Division of Veterinary Pharmacy, Pharmacology and Toxicology, Utrecht University, Utrecht, The Netherlands; Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Saskia Braber
- Division of Veterinary Pharmacy, Pharmacology and Toxicology, Utrecht University, Utrecht, The Netherlands
| | - Peyman Akbari
- Division of Veterinary Pharmacy, Pharmacology and Toxicology, Utrecht University, Utrecht, The Netherlands; Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands; Nutricia Research, Utrecht, The Netherlands
| | - Johanna Fink-Gremmels
- Division of Veterinary Pharmacy, Pharmacology and Toxicology, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
87
|
Kojibiose ameliorates arachidic acid-induced metabolic alterations in hyperglycaemic rats. Br J Nutr 2015; 114:1395-402. [DOI: 10.1017/s0007114515003153] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
AbstractHerein we hypothesise the positive effects of kojibiose (KJ), a prebiotic disaccharide, selected for reducing hepatic expression of inflammatory markers in vivo that could modulate the severity of saturated arachidic acid (ARa)-induced liver dysfunction in hyperglycaemic rats. Animals were fed daily (20 d) with ARa (0·3 mg) together or not with KJ (22 mg approximately 0·5 %, w/w diet). Glucose, total TAG and cholesterol contents and the phospholipid profile were determined in serum samples. Liver sections were collected for the expression (mRNA) of enzymes and innate biomarkers, and intrahepatic macrophage and T-cell populations were analysed by flow cytometry. ARa administration increased the proportion of liver to body weight that was associated with an increased (by 11 %) intrahepatic macrophage population. These effects were ameliorated when feeding with KJ, which also normalised the plasmatic levels of TAG and N-acyl-phosphatidylethenolamine in response to tissue damage. These results indicate that daily supplementation of KJ significantly improves the severity of ARa-induced hepatic alterations.
Collapse
|
88
|
Verheijden KAT, Akbari P, Willemsen LEM, Kraneveld AD, Folkerts G, Garssen J, Fink-Gremmels J, Braber S. Inflammation-induced expression of the alarmin interleukin 33 can be suppressed by galacto-oligosaccharides. Int Arch Allergy Immunol 2015; 167:127-36. [PMID: 26304032 DOI: 10.1159/000437327] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 06/30/2015] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The alarmin interleukin 33 (IL-33) and its receptor ST2 play an important role in mucosal barrier tissues, and seem to be crucial for Th2-cell mediated host defense. Galacto-oligosaccharides (GOS), used in infant formulas, exhibit gut and immune modulatory effects. To enhance our understanding of the immunomodulatory capacity of GOS, this study investigated the impact of dietary GOS intervention on IL-33 and ST2 expression related to intestinal barrier dysfunction and asthma. METHODS B6C3F1 and BALB/c mice were fed a control diet with or without 1% GOS. To simulate intestinal barrier dysfunction, B6C3F1 mice received a gavage with the mycotoxin deoxynivalenol (DON). To mimic asthma-like inflammatory airway responses, BALB/c mice were sensitized on day 0 and challenged on days 7-11 with house-dust mite (HDM) allergen. Samples from the intestines and lungs were collected for IL-33 and ST2 analysis by qRT-PCR, immunoblotting and immunohistochemistry. RESULTS Dietary GOS counteracted the DON-induced IL-33 mRNA expression and changed the IL-33 distribution pattern in the mouse small intestine. The IL-33 mRNA expression was positively correlated to the intestinal permeability. A strong positive correlation was also observed between IL-33 mRNA expression in the lung and the number of bronchoalveolar fluid cells. Reduced levels of IL-33 protein, altered IL-33 distribution and reduced ST2 mRNA expression were observed in the lungs of HDM-allergic mice after GOS intervention. CONCLUSIONS Dietary GOS mitigated IL-33 at the mucosal surfaces in a murine model for intestinal barrier dysfunction and HDM-induced asthma. This promising effect may open up new avenues to use GOS not only as a prebiotic in infant nutrition, but also as a functional ingredient that targets inflammatory processes and allergies associated with IL-33 expression.
Collapse
Affiliation(s)
- Kim A T Verheijden
- Division of Veterinary Pharmacy, Pharmacology and Toxicology, Utrecht University, Utrecht, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
89
|
Altmann K, Wutkowski A, Kämpfer S, Klempt M, Lorenzen PC, Clawin-Rädecker I. Comparison of the efficiency of different NF membranes for the enrichment of milk oligosaccharides from bovine milk. Eur Food Res Technol 2015. [DOI: 10.1007/s00217-015-2505-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
90
|
Capitán-Cañadas F, Ocón B, Aranda CJ, Anzola A, Suárez MD, Zarzuelo A, de Medina FS, Martínez-Augustin O. Fructooligosaccharides exert intestinal anti-inflammatory activity in the CD4+ CD62L+ T cell transfer model of colitis in C57BL/6J mice. Eur J Nutr 2015; 55:1445-54. [PMID: 26154776 DOI: 10.1007/s00394-015-0962-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 06/10/2015] [Indexed: 12/27/2022]
Abstract
PURPOSE Fructooligosaccharides (FOS) are used as functional foods due to their prebiotic effects. Intestinal anti-inflammatory activity has been established in most, but not all, studies in animal models of colitis, using mainly chemically induced inflammation. Our goal was to test the effect of FOS (degree of polymerization 2-8) in the chronic, lymphocyte-driven CD4+ CD62L+ T cell transfer model of colitis. METHODS Colitis was induced by transfer of CD4+ CD62L+ T cells to C57BL/6J Rag1(-/-) mice. FOS (75 mg day(-1)) was administered by gavage as a post-treatment. Three groups were established: non-colitic (NC), colitic control (C, CD4+ CD62L+ transferred mice treated with vehicle) and colitic+FOS (C+FOS, similar but treated with FOS). Mice were killed after 13 days. RESULTS Treatment of mice with FOS ameliorated colitis, as evidenced by an increase in body weight, a lesser myeloperoxidase and alkaline phosphatase activities, a lower secretion of proinflammatory cytokines by mesenteric lymph node cells ex vivo (IFN-γ, IL-17, and TNF-α), and a higher colonic expression of occludin (C+FOS vs. C, p < 0.05). Increased relative abundance of lactic acid bacteria was observed in FOS-treated mice (p < 0.05). CONCLUSIONS FOS exert intestinal anti-inflammatory activity in T lymphocyte-dependent colitis, suggesting it may be useful in the management of inflammatory bowel disease in appropriate conditions.
Collapse
Affiliation(s)
- Fermín Capitán-Cañadas
- Department of Biochemistry and Molecular Biology II, CIBERehd, School of Pharmacy, University of Granada, Campus de Cartuja s/n, C.P. 18071, Granada, Spain
| | - Borja Ocón
- Department of Pharmacology, CIBERehd, School of Pharmacy, University of Granada, Campus de Cartuja s/n, C.P. 18071, Granada, Spain
| | - Carlos José Aranda
- Department of Biochemistry and Molecular Biology II, CIBERehd, School of Pharmacy, University of Granada, Campus de Cartuja s/n, C.P. 18071, Granada, Spain
| | - Andrea Anzola
- Department of Pharmacology, CIBERehd, School of Pharmacy, University of Granada, Campus de Cartuja s/n, C.P. 18071, Granada, Spain
| | - María Dolores Suárez
- Department of Biochemistry and Molecular Biology II, CIBERehd, School of Pharmacy, University of Granada, Campus de Cartuja s/n, C.P. 18071, Granada, Spain
| | - Antonio Zarzuelo
- Department of Pharmacology, CIBERehd, School of Pharmacy, University of Granada, Campus de Cartuja s/n, C.P. 18071, Granada, Spain
| | - Fermín Sánchez de Medina
- Department of Pharmacology, CIBERehd, School of Pharmacy, University of Granada, Campus de Cartuja s/n, C.P. 18071, Granada, Spain
| | - Olga Martínez-Augustin
- Department of Biochemistry and Molecular Biology II, CIBERehd, School of Pharmacy, University of Granada, Campus de Cartuja s/n, C.P. 18071, Granada, Spain.
| |
Collapse
|
91
|
Lehmann S, Hiller J, van Bergenhenegouwen J, Knippels LMJ, Garssen J, Traidl-Hoffmann C. In Vitro Evidence for Immune-Modulatory Properties of Non-Digestible Oligosaccharides: Direct Effect on Human Monocyte Derived Dendritic Cells. PLoS One 2015; 10:e0132304. [PMID: 26148091 PMCID: PMC4493044 DOI: 10.1371/journal.pone.0132304] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 06/11/2015] [Indexed: 11/19/2022] Open
Abstract
Infant formulas containing non-digestible oligosaccharides (NDO) similar to the composition in breast milk or a combination of lactic acid bacteria (LAB) and NDO have been shown to harbor preventive effects towards immune-regulatory disorders. The aim of this study was to investigate the immune-modulatory potential of non-digestible short chain galacto- and long chain fructo-oligosaccharides (scGOS/lcFOS) mimicking the natural distribution of oligosaccharides in human breast milk in presence or absence of certain LAB strains in human monocyte derived dendritic cells (MoDC). Immature human MoDC prepared from peripheral blood of healthy non-atopic volunteers were screened in vitro after stimulation with specific scGOS/lcFOS in presence or absence of LAB. IL-10 and IL-12p70 release was analyzed after 24 hours in cell-free supernatants by enzyme-linked immunosorbent assay (ELISA). A luminex-based assay was conducted to assess further cytokine and chemokine release by MoDC. To investigate the resulting T cell response, stimulated MoDC were co-incubated with naïve T cells in allogeneic stimulation assays and intracellular Foxp3 expression, as well as immune-suppressive capacity was determined. Oligosaccharides did not induce relevant amounts of IL-12p70 production, but did promote IL-10 release by MoDC. Furthermore, scGOS/lcFOS mixtures exerted a significant enhancing effect on LAB induced IL-10 secretion by MoDC while no increase in IL-12p70 production was observed. Blocking toll like receptor (TLR)4 abrogated the increase in IL-10 in both the direct stimulation and the LAB stimulation of MoDC, suggesting that scGOS/lcFOS act via TLR4. Finally, scGOS/lcFOS-treated MoDC were shown to upregulate the number of functional suppressive Foxp3 positive T cells following allogeneic stimulation. Our results indicate anti-inflammatory and direct, microbiota independent, immune-modulatory properties of scGOS/lcFOS mixtures on human MoDC suggesting a possible induction of regulatory T cells (Tregs). The tested combinations of LAB and scGOS/lcFOS might represent a useful dietary ingredient for the maintenance of intestinal homeostasis via the induction of Tregs.
Collapse
Affiliation(s)
- Sarah Lehmann
- Institute of Environmental Medicine, UNIKA-T, Technische Universität München, Augsburg, Germany
- Center of Allergy and Environment (ZAUM), Member of the German Center for Lung Research (DZL), Technische Universität & Helmholtz Zentrum München, Munich, Germany
| | - Julia Hiller
- Institute of Environmental Medicine, UNIKA-T, Technische Universität München, Augsburg, Germany
- Center of Allergy and Environment (ZAUM), Member of the German Center for Lung Research (DZL), Technische Universität & Helmholtz Zentrum München, Munich, Germany
| | - Jeroen van Bergenhenegouwen
- Nutricia Research, Department of Immunology, Utrecht, The Netherlands
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Leon M. J. Knippels
- Nutricia Research, Department of Immunology, Utrecht, The Netherlands
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Johan Garssen
- Nutricia Research, Department of Immunology, Utrecht, The Netherlands
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Claudia Traidl-Hoffmann
- Institute of Environmental Medicine, UNIKA-T, Technische Universität München, Augsburg, Germany
- CK CARE-Christine Kühne Center for Allergy Research and Education, Davos, Switzerland
| |
Collapse
|
92
|
Akbari P, Braber S, Alizadeh A, Verheijden KAT, Schoterman MHC, Kraneveld AD, Garssen J, Fink-Gremmels J. Galacto-oligosaccharides Protect the Intestinal Barrier by Maintaining the Tight Junction Network and Modulating the Inflammatory Responses after a Challenge with the Mycotoxin Deoxynivalenol in Human Caco-2 Cell Monolayers and B6C3F1 Mice. J Nutr 2015; 145:1604-13. [PMID: 26019243 DOI: 10.3945/jn.114.209486] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 05/08/2015] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND The integrity of the epithelial layer in the gastrointestinal tract protects organisms from exposure to luminal antigens, which are considered the primary cause of chronic intestinal inflammation and allergic responses. The common wheat-associated fungal toxin deoxynivalenol acts as a specific disruptor of the intestinal tight junction network and hence might contribute to the pathogenesis of inflammatory bowel diseases. OBJECTIVE The aim of the current study was to assess whether defined galacto-oligosaccharides (GOSs) can prevent deoxynivalenol-induced epithelial dysfunction. METHODS Human epithelial intestinal Caco-2 cells, pretreated with different concentrations of GOSs (0.5%, 1%, and 2%) for 24 h, were stimulated with 4.2-μM deoxynivalenol (24 h), and 6/7-wk-old male B6C3F1 mice were fed a diet supplemented with 1% GOSs for 2 wk before being orally exposed to deoxynivalenol (25 mg/kg body weight, 6 h). Barrier integrity was determined by measuring transepithelial electrical resistance (TEER) and intestinal permeability to marker molecules. A calcium switch assay was conducted to study the assembly of epithelial tight junction proteins. Alterations in tight junction and cytokine expression were assessed by quantitative reverse transcriptase-polymerase chain reaction, Western blot analysis, or ELISA, and their localization was visualized by immunofluorescence microscopy. Sections of the proximal and distal small intestine were stained with hematoxylin/eosin for histomorphometric analysis. RESULTS The in vitro data showed that medium supplemented with 2% GOSs improved tight junction assembly reaching an acceleration of 85% after 6 h (P < 0.05). In turn, GOSs prevented the deoxynivalenol-induced loss of epithelial barrier function as measured by TEER (114% of control), and paracellular flux of Lucifer yellow (82.7% of prechallenge values, P < 0.05). Moreover, GOSs stabilized the expression and cellular distribution of claudin3 and suppressed by >50% the deoxynivalenol-induced synthesis and release of interleukin-8 [IL8/chemokine CXC motif ligand (CXCL8)] (P < 0.05). In mice, GOSs prevented the deoxynivalenol-induced mRNA overexpression of claudin3 (P = 0.022) and CXCL8 homolog keratinocyte hemoattractant (Kc) (Cxcl1) (P = 0.06) as well as the deoxynivalenol-induced morphologic defects. CONCLUSIONS The results demonstrate that GOSs stimulate the tight junction assembly and in turn mitigate the deleterious effects of deoxynivalenol on the intestinal barrier of Caco-2 cells and on villus architecture of B6C3F1 mice.
Collapse
Affiliation(s)
- Peyman Akbari
- Divisions of Veterinary Pharmacy, Pharmacology, and Toxicology, and Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Saskia Braber
- Divisions of Veterinary Pharmacy, Pharmacology, and Toxicology, and
| | - Arash Alizadeh
- Divisions of Veterinary Pharmacy, Pharmacology, and Toxicology, and Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Kim A T Verheijden
- Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | | | - Aletta D Kraneveld
- Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Johan Garssen
- Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands; Nutricia Research, Utrecht, The Netherlands
| | | |
Collapse
|
93
|
Intrinsic immunomodulatory effects of low-digestible carbohydrates selectively extend their anti-inflammatory prebiotic potentials. BIOMED RESEARCH INTERNATIONAL 2015; 2015:162398. [PMID: 25977916 PMCID: PMC4419225 DOI: 10.1155/2015/162398] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 09/17/2014] [Accepted: 09/17/2014] [Indexed: 12/21/2022]
Abstract
The beneficial effects of carbohydrate-derived fibers are mainly attributed to modulation of the microbiota, increased colonic fermentation, and the production of short-chain fatty acids. We studied the direct immune responses to alimentary fibers in in vitro and in vivo models. Firstly, we evaluated the immunomodulation induced by nine different types of low-digestible fibers on human peripheral blood mononuclear cells. None of the fibers tested induced cytokine production in baseline conditions. However, only one from all fibers almost completely inhibited the production of anti- and proinflammatory cytokines induced by bacteria. Secondly, the impact of short- (five days) and long-term (three weeks) oral treatments with selected fibers was assessed in the trinitrobenzene-sulfonic acid colitis model in mice. The immunosuppressive fiber significantly reduced levels of inflammatory markers over both treatment periods, whereas a nonimmunomodulatory fiber had no effect. The two fibers did not differ in terms of the observed fermentation products and colonic microbiota after three weeks of treatment, suggesting that the anti-inflammatory action was not related to prebiotic properties. Hence, we observed a direct effect of a specific fiber on the murine immune system. This intrinsic, fiber-dependent immunomodulatory potential may extend prebiotic-mediated protection in inflammatory bowel disease.
Collapse
|
94
|
Bermudez-Brito M, Sahasrabudhe NM, Rösch C, Schols HA, Faas MM, de Vos P. The impact of dietary fibers on dendritic cell responses in vitro is dependent on the differential effects of the fibers on intestinal epithelial cells. Mol Nutr Food Res 2015; 59:698-710. [PMID: 25620425 DOI: 10.1002/mnfr.201400811] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Revised: 01/15/2015] [Accepted: 01/16/2015] [Indexed: 12/19/2022]
Abstract
SCOPE In the present study, the direct interaction of commonly consumed fibers with epithelial or dendritic cells (DCs) was studied. METHODS AND RESULTS The fibers were characterized for their sugar composition and chain length profile. When in direct contact, fibers activate DCs only mildly. This was different when DCs and fibers were co-cultured together with supernatants from human epithelial cells (Caco spent medium). Caco spent medium enhanced the production of IL-12, IL-1Ra, IL-6, IL-8, TNF-α, MCP-1 (monocyte chemotactic protein), and MIP-1α but this was strongly attenuated by the dietary fibers. This attenuating effect on proinflammatory cytokines was dependent on the interaction of the fibers with Toll-like receptors as it was reduced by Pepinh-myd88. The interaction of galacto-oligosaccharides, chicory inulin, wheat arabinoxylan, barley β-glucan with epithelial cells and DCs led to changes in the production of the Th1 cytokines in autologous T cells, while chicory inulin, and barley β-glucan reduced the Th2 cytokine IL-6. The Treg-promoting cytokine IL-10 was induced by galacto-oligosaccharides whereas chicory inulin decreased the IL-10 production. CONCLUSIONS Our results suggest that dietary fibers can modulate the host immune system not only by the recognized mechanism of effects on microbiota but also by direct interaction with the consumer's mucosa. This modulation is dietary fiber type dependent.
Collapse
Affiliation(s)
- Miriam Bermudez-Brito
- Top Institute Food and Nutrition, Wageningen, The Netherlands; Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | | | | | | | | | | |
Collapse
|
95
|
Bhatia S, Prabhu PN, Benefiel AC, Miller MJ, Chow J, Davis SR, Gaskins HR. Galacto-oligosaccharides may directly enhance intestinal barrier function through the modulation of goblet cells. Mol Nutr Food Res 2015; 59:566-73. [PMID: 25421108 DOI: 10.1002/mnfr.201400639] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Revised: 11/04/2014] [Accepted: 11/06/2014] [Indexed: 12/24/2022]
Abstract
SCOPE Here we have tested the hypothesis that prebiotic galacto-oligosaccharides (GOS) may enhance mucosal barrier function through direct modulation of goblet cell function. METHODS AND RESULTS Human adenocarcinoma-derived LS174T cells, which exhibit an intestinal goblet cell-like phenotype, were used to examine the non-prebiotic effects of GOS on goblet cell functions. LS174T cells were treated with GOS, and the expression of goblet cell secretory product genes mucin 2 (MUC2), trefoil factor 3 (TFF3), resistin-like molecule beta (RETNLB) and the Golgi-sulfotransferase genes, carbohydrate (N-acetylglucosamine-6-O) sulfotransferase 5 (CHST5) and galactose-3-O-sulfotransferase 2 (GAL3ST2), was determined by real-time quantitative RT-PCR. In addition, the abundance of CHST5, TFF3 and RETNLB was confirmed by Western blot analysis. Following treatment with GOS for 72 h, the expression of MUC2 was significantly upregulated 2-4-fold, CHST5 and RETNLB, 5-7-fold, and TFF3 2-4-fold. Western blot analysis demonstrated increased abundance of RETNLB, TFF3 and CHST5. Addition of the Th2 cytokine IL-13 along with GOS resulted in synergistic induction of RETNLB and CHST5. IL-8 secretion was not affected by GOS treatment, suggesting that the effects of GOS are not mediated through an inflammatory pathway. CONCLUSION Collectively, the data indicate that GOS may enhance mucosal barrier function through direct stimulation of intestinal goblet cells.
Collapse
Affiliation(s)
- Shikha Bhatia
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, IL, USA
| | | | | | | | | | | | | |
Collapse
|
96
|
Vendrig JC, Coffeng LE, Fink-Gremmels J. Effects of orally administered galacto-oligosaccharides on immunological parameters in foals: a pilot study. BMC Vet Res 2014; 10:278. [PMID: 25407340 PMCID: PMC4241218 DOI: 10.1186/s12917-014-0278-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 11/10/2014] [Indexed: 11/10/2022] Open
Abstract
Background In the first phase of life, in which the immune system is primed and the bacterial colonization of epithelial surfaces takes place, foals are highly susceptible to bacterial infections. Next to strategies to optimize maternally acquired immunity in individual foals, current research explores other options to modulate immune responses in foals. During the past decades, oligosaccharide supplements were developed to mimic beneficial properties of the oligosaccharides, which are present in colostrum and milk. In human infants and laboratory animal species, dietary supplementation with galacto-oligosaccharides (GOS) has been shown to result in prebiotic and immunomodulating effects, with long-term beneficial consequences for both defensive and allergic immune responses. As yet, no studies are published concerning the in vivo effects of GOS in horses. The current study was designed as a pilot study to investigate the effects of an orally applied, commercially available GOS product in a group of pony foals. The treatment and the control group consisted of six and four foals, respectively. Foals were treated during the first four weeks of life and subsequently followed up for another ten weeks. Results In peripheral blood mononuclear cells (PBMCs) derived from GOS-treated foals at day 28, a standardized lipopolysaccharide challenge resulted in significantly lower relative mRNA expression levels of the pro-inflammatory cytokines interferon-γ and interleukin-6 compared with PBMCs of control foals. In the 98-day period of investigation, no significant effects of the GOS supplement were observed on clinical and blood parameters for immunity and general health in these foals. Conclusions Based on these first results, we can conclude that this dose regimen of GOS was well accepted by the foals and did not result in any detectable undesirable side effects. More clinical trials are required to confirm the attenuating effects of GOS treatment on equine pro-inflammatory immune responses and to implement this into practice. Electronic supplementary material The online version of this article (doi:10.1186/s12917-014-0278-4) contains supplementary material, which is available to authorized users.
Collapse
|
97
|
Murofushi Y, Villena J, Morie K, Kanmani P, Tohno M, Shimazu T, Aso H, Suda Y, Hashiguchi K, Saito T, Kitazawa H. The toll-like receptor family protein RP105/MD1 complex is involved in the immunoregulatory effect of exopolysaccharides from Lactobacillus plantarum N14. Mol Immunol 2014; 64:63-75. [PMID: 25466614 DOI: 10.1016/j.molimm.2014.10.027] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Revised: 10/28/2014] [Accepted: 10/29/2014] [Indexed: 01/10/2023]
Abstract
The radioprotective 105 (RP105)/MD1 complex is a member of the Toll-like receptor (TLR) family. It was reported that RP105/MD1 cooperates with the lipopolysaccharide (LPS) receptor TLR4/MD2 complex and plays a crucial role in the response of immune cells to LPS. This work evaluated whether RP105, TLR4 or TLR2 were involved in the immunoregulatory capacities of Lactobacillus plantarum N14 (LP14) or its exopolysaccharides (EPS). EPS from LP14 were fractionated into neutral (NPS) and acidic (APS) EPS by anion exchange chromatography. Experiments with transfectant HEK(RP105/MD1) and HEK(TLR2) cells demonstrated that LP14 strongly activated NF-κB via RP105 and TLR2. When we studied the capacity of APS to activate NF-κB pathway in HEK(RP105/MD1) and HEK(TLR4) cells; we observed that APS strongly stimulated both transfectant cells. Our results also showed that LP14 and APS were able to decrease the production of pro-inflammatory cytokines (IL-6, IL-8 and MCP-1) in porcine intestinal epithelial (PIE) cells in response to enterotoxigenic Escherichia coli (ETEC) challenge. In order to confirm the role of TLR2, TLR4 and RP105 in the immunoregulatory effect of APS from LP14, we used small interfering RNA (siRNA) to knockdown these receptors in PIE cells. The capacity of LP14 and APS to modulate pro-inflammatory cytokine expression was significantly reduced in PIE(RP105-/-) cells. It was also shown that LP14 and APS were capable of upregulating negative regulators of the TLR signaling in PIE cells. This work describes for the first time that a Lactobacillus strain and its EPS reduce inflammation in intestinal epithelial cells in a RP105/MD1-dependend manner.
Collapse
Affiliation(s)
- Yo Murofushi
- Food and Feed Immunology Group, Graduate School of Agricultural Science, Tohoku University, Aobaku, Sendai 981-8555, Japan.
| | - Julio Villena
- Food and Feed Immunology Group, Graduate School of Agricultural Science, Tohoku University, Aobaku, Sendai 981-8555, Japan; Laboratory of Immunobiotechnology, CERELA-CONICET, Tucuman, Argentina.
| | - Kyoko Morie
- Food and Feed Immunology Group, Graduate School of Agricultural Science, Tohoku University, Aobaku, Sendai 981-8555, Japan.
| | - Paulraj Kanmani
- Food and Feed Immunology Group, Graduate School of Agricultural Science, Tohoku University, Aobaku, Sendai 981-8555, Japan.
| | - Masanori Tohno
- Functional Feed Research Team, National Institute of Livestock and Grassland Science, National Agricultural Research Organization, Nasushiobara, Tochigi 329-2793, Japan.
| | - Tomoyuki Shimazu
- Laboratory of Animal Breeding and Genetics, Graduate School of Agricultural Science, Tohoku University, Aobaku, Sendai 981-8555, Japan.
| | - Hisashi Aso
- Cell Biology Laboratory, Graduate School of Agricultural Science, Tohoku University, Aobaku, Sendai 981-8555, Japan.
| | - Yoshihito Suda
- Department of Food, Agriculture and Environment, Miyagi University, Sendai 982-0215, Japan.
| | | | - Tadao Saito
- Food and Feed Immunology Group, Graduate School of Agricultural Science, Tohoku University, Aobaku, Sendai 981-8555, Japan.
| | - Haruki Kitazawa
- Food and Feed Immunology Group, Graduate School of Agricultural Science, Tohoku University, Aobaku, Sendai 981-8555, Japan.
| |
Collapse
|
98
|
Wachi S, Kanmani P, Tomosada Y, Kobayashi H, Yuri T, Egusa S, Shimazu T, Suda Y, Aso H, Sugawara M, Saito T, Mishima T, Villena J, Kitazawa H. Lactobacillus delbrueckii TUA4408L and its extracellular polysaccharides attenuate enterotoxigenic Escherichia coli-induced inflammatory response in porcine intestinal epitheliocytes via Toll-like receptor-2 and 4. Mol Nutr Food Res 2014; 58:2080-93. [PMID: 24995380 DOI: 10.1002/mnfr.201400218] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Revised: 05/31/2014] [Accepted: 06/27/2014] [Indexed: 01/04/2023]
Abstract
SCOPE Immunobiotics are known to modulate intestinal immune responses by regulating Toll-like receptor (TLR) signaling pathways, which are responsible for the induction of cytokines and chemokines in response to microbial-associated molecular patterns. However, little is known about the immunomodulatory activity of compounds or molecules from immunobiotics. METHODS AND RESULTS We evaluated whether Lactobacillus delbrueckii subsp. delbrueckii TUA4408L (Ld) or its extracellular polysaccharide (EPS): acidic EPS (APS) and neutral EPS (NPS), modulated the response of porcine intestinal epitheliocyte (PIE) cells against Enterotoxigenic Escherichia coli (ETEC) 987P. The roles of TLR2, TLR4, and TLR negative regulators in the immunoregulatory effects were also studied. ETEC-induced inflammatory cytokines were downregulated when PIE cells were prestimulated with both Ld or EPSs. Ld, APS, and NPS inhibited ETEC mediated mitogen-activated protein kinase (MAPK) and nuclear factor-κB (NF-κB) activation by upregulating TLR negative regulators. The capability of Ld to suppress inflammatory cytokines was diminished when PIE cells were blocked with anti-TLR2 antibody, while APS failed to suppress inflammatory cytokines when cells were treated with anti-TLR4 antibody. Induction of Ca²⁺ fluxes in TLR knockdown cells confirmed that TLR2 plays a principal role in the immunomodulatory action of Ld, while the activity of APS is mediated by TLR4. In addition, NPS activity depends on both TLR4 and TLR2. CONCLUSION Ld and its EPS have the potential to be used for the development of anti-inflammatory functional foods to prevent intestinal diseases in both humans and animals.
Collapse
Affiliation(s)
- Satoshi Wachi
- Food and Feed Immunology Group, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
99
|
|
100
|
Capitán-Cañadas F, Ortega-González M, Guadix E, Zarzuelo A, Suárez MD, de Medina FS, Martínez-Augustin O. Prebiotic oligosaccharides directly modulate proinflammatory cytokine production in monocytes via activation of TLR4. Mol Nutr Food Res 2013; 58:1098-110. [PMID: 24549946 DOI: 10.1002/mnfr.201300497] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Revised: 09/10/2013] [Accepted: 10/12/2013] [Indexed: 11/06/2022]
Abstract
SCOPE Prebiotic oligosaccharides are currently used in a variety of clinical settings for their effects on intestinal microbiota. Here, we have examined the direct, microbiota independent, effects of prebiotics on monocytes and T lymphocytes in vitro. METHODS AND RESULTS Prebiotics generally evoked cytokine secretion (TNF-α, IL-6, and IL-10) by mouse splenocytes but inhibited LPS -induced IFN-γ and IL-17 release. Inulin was found to enhance LPS-induced IL-10 secretion. Splenocytes from TLR4(-/-) (where TLR is Toll-like receptor) mice showed a markedly depressed response. Conversely, in both basal and LPS-stimulated conditions, prebiotic inhibition of IFN-γ levels was preserved. These results suggested a predominant effect on monocytes via TLR4 ligation and possible inhibition of T cells. Hence, we studied the modulation of primary rat monocytes and T lymphocytes, focusing on fructooligosaccharides (FOS) and inulin. In monocytes, FOS and inulin induced TNF-α, growth-regulated oncogene α, and IL-10, but not IL-1β release. The NF-κB inhibitor Bay 11-7082 fully prevented these effects. Pharmacological evidence also indicated a significant involvement of mitogen-activated protein kinase and phosphatidylinositol-3-kinase. There was little effect on T cells. FOS and inulin also generally increased TNF-α, IL-1β, and IL-10, but not IL-8, in human peripheral blood monocytes. CONCLUSION We conclude that prebiotics may act as TLR4 ligands or as indirect TLR4 modulators to upregulate cytokine secretion in monocytes.
Collapse
Affiliation(s)
- Fermín Capitán-Cañadas
- Department of Biochemistry and Molecular Biology II, CIBERehd, School of Pharmacy, University of Granada, Granada, Spain
| | | | | | | | | | | | | |
Collapse
|