1
|
Dmytriv TR, Storey KB, Lushchak VI. Intestinal barrier permeability: the influence of gut microbiota, nutrition, and exercise. Front Physiol 2024; 15:1380713. [PMID: 39040079 PMCID: PMC11260943 DOI: 10.3389/fphys.2024.1380713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 05/29/2024] [Indexed: 07/24/2024] Open
Abstract
The intestinal wall is a selectively permeable barrier between the content of the intestinal lumen and the internal environment of the body. Disturbances of intestinal wall permeability can potentially lead to unwanted activation of the enteric immune system due to excessive contact with gut microbiota and its components, and the development of endotoxemia, when the level of bacterial lipopolysaccharides increases in the blood, causing chronic low-intensity inflammation. In this review, the following aspects are covered: the structure of the intestinal wall barrier; the influence of the gut microbiota on the permeability of the intestinal wall via the regulation of functioning of tight junction proteins, synthesis/degradation of mucus and antioxidant effects; the molecular mechanisms of activation of the pro-inflammatory response caused by bacterial invasion through the TLR4-induced TIRAP/MyD88 and TRAM/TRIF signaling cascades; the influence of nutrition on intestinal permeability, and the influence of exercise with an emphasis on exercise-induced heat stress and hypoxia. Overall, this review provides some insight into how to prevent excessive intestinal barrier permeability and the associated inflammatory processes involved in many if not most pathologies. Some diets and physical exercise are supposed to be non-pharmacological approaches to maintain the integrity of intestinal barrier function and provide its efficient operation. However, at an early age, the increased intestinal permeability has a hormetic effect and contributes to the development of the immune system.
Collapse
Affiliation(s)
- Tetiana R. Dmytriv
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, Ivano-Frankivsk, Ukraine
- Research and Development University, Ivano-Frankivsk, Ukraine
| | | | - Volodymyr I. Lushchak
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, Ivano-Frankivsk, Ukraine
- Research and Development University, Ivano-Frankivsk, Ukraine
| |
Collapse
|
2
|
Simon DA, Kellermayer R. INVITED COMMENTARY on Andersen S, et al. Developmental Windows of Environmental Vulnerability for Inflammatory Bowel Disease. JOURNAL OF PEDIATRICS. CLINICAL PRACTICE 2024; 11:200104. [PMID: 38827481 PMCID: PMC11138252 DOI: 10.1016/j.jpedcp.2024.200104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 02/21/2024] [Indexed: 06/04/2024]
Affiliation(s)
- David A. Simon
- Division of Pediatric Gastroenterology, Texas Children’s Hospital Baylor College of Medicine, Houston, TX
| | - Richard Kellermayer
- Division of Pediatric Gastroenterology, Texas Children’s Hospital Baylor College of Medicine, Houston, TX
- USDA/ARS Children’s Nutrition Research Center, Houston, TX
| |
Collapse
|
3
|
Li G, Wu X, Gao X, Lin R, Chen L, Sun M, Jia J, Liu Z, Fang L, Wu W. Long-term exclusive enteral nutrition remodels gut microbiota and alleviates TNBS-induced colitis in mice. Food Funct 2022; 13:1725-1740. [PMID: 35085377 DOI: 10.1039/d1fo03579g] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Exclusive enteral nutrition (EEN) provides an effective strategy for the induction of clinical remission in pediatric Crohn’s disease. However, the feasibility of long-term EEN in management of disease and the...
Collapse
Affiliation(s)
- Gengfeng Li
- Center for IBD Research, Department of Gastroenterology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 301 Yanchang Road, Shanghai 200072, China.
| | - Xiaohan Wu
- Center for IBD Research, Department of Gastroenterology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 301 Yanchang Road, Shanghai 200072, China.
| | - Xiang Gao
- Center for IBD Research, Department of Gastroenterology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 301 Yanchang Road, Shanghai 200072, China.
| | - Ritian Lin
- Center for IBD Research, Department of Gastroenterology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 301 Yanchang Road, Shanghai 200072, China.
| | - Liang Chen
- Center for IBD Research, Department of Gastroenterology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 301 Yanchang Road, Shanghai 200072, China.
| | - Mingming Sun
- Center for IBD Research, Department of Gastroenterology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 301 Yanchang Road, Shanghai 200072, China.
| | - Junwan Jia
- Center for IBD Research, Department of Gastroenterology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 301 Yanchang Road, Shanghai 200072, China.
| | - Zhanju Liu
- Center for IBD Research, Department of Gastroenterology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 301 Yanchang Road, Shanghai 200072, China.
| | - Leilei Fang
- Center for IBD Research, Department of Gastroenterology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 301 Yanchang Road, Shanghai 200072, China.
| | - Wei Wu
- Center for IBD Research, Department of Gastroenterology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 301 Yanchang Road, Shanghai 200072, China.
| |
Collapse
|
4
|
Jang S, Kim Y, Lee C, Kwon B, Noh J, Jee JJ, Yoon SS, Koh H, Park S. The Effect of Formula-based Nutritional Treatment on Colitis in a Murine Model. J Korean Med Sci 2021; 36:e342. [PMID: 34962114 PMCID: PMC8728592 DOI: 10.3346/jkms.2021.36.e342] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 10/29/2021] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Exclusive enteral nutrition (EEN) induces remission in pediatric Crohn's disease (CD). The exact mechanism of EEN therapy in CD is unknown, but alteration of the intestinal microflora after EEN is thought to affect mucosal healing. To determine the link between EEN therapy and therapeutic efficacy in CD, we established a murine model of dextran sulfate sodium (DSS)-induced colitis and applied EEN therapy. METHODS Eight-week-old C57BL/6 mice were administered DSS for 4 days to induce colitis, and either normal chow (NC) or EEN was administered for the following 4 days. The mice were grouped according to the feeding pattern after DSS administration: DSS/NC and DSS/EEN groups. The clinical course was confirmed via daily observation of the weight and stool. Fecal samples were collected and 16sRNA sequencing was used. The mice were sacrificed to confirm colonic histopathology. RESULTS Weight reduction and increase in disease activity were observed as the day progressed for 4 days after DSS administration. There was significant weight recovery and improvement in disease activity in the EEN group compared to that in the NC group. Verrucomicrobia and Proteobacteria abundances tended to increase and Bacteroidetes abundance decreased in the EEN group. In the EEN group, significant changes in the β-diversity of the microbiota were observed. In the analysis of microbiome species, abundances of Akkermansia muciniphila, Clostridium cocleatum, mucin-degrading bacteria, Flintibacter butyricus, and Parabacteroides goldsteinii, which are beneficial microbiota, were significantly increased in the EEN group compared to those in the NC group. More abundant mucins were confirmed in the colonic histopathology of the EEN group. These microbial and histopathological differences suggested that EEN might improve colitis symptoms in a murine colitis model by promoting mucin recycling and subsequently inducing the healing effect of the gut barrier. CONCLUSION EEN showed clinical efficacy in a murine model of colitis. Based on the increase in mucin-degrading bacteria and the pathological increase in mucin production after EEN administration, it can be observed that mucin plays an important role in the therapeutic effect of EEN.
Collapse
Affiliation(s)
- Sooyoung Jang
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Yonsei University College of Medicine, Severance Fecal Microbiota Transplantation Center, Severance Hospital, Seoul, Korea
| | - Younjuong Kim
- Department of Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Changjun Lee
- Department of Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Bomi Kwon
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Yonsei University College of Medicine, Severance Fecal Microbiota Transplantation Center, Severance Hospital, Seoul, Korea
| | - Jihye Noh
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Yonsei University College of Medicine, Severance Fecal Microbiota Transplantation Center, Severance Hospital, Seoul, Korea
| | - Jai J Jee
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Yonsei University College of Medicine, Severance Fecal Microbiota Transplantation Center, Severance Hospital, Seoul, Korea
| | - Sang Sun Yoon
- Department of Microbiology and Immunology, Brain Korea 21 Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Korea
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, Korea
| | - Hong Koh
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Yonsei University College of Medicine, Severance Fecal Microbiota Transplantation Center, Severance Hospital, Seoul, Korea
| | - Sowon Park
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Yonsei University College of Medicine, Severance Fecal Microbiota Transplantation Center, Severance Hospital, Seoul, Korea.
| |
Collapse
|
5
|
Krishna M, Salako A, Fofanova T, Kellermayer R. Parental Education May Differentially Impact Pediatric Inflammatory Bowel Disease Phenotype Risk. Inflamm Bowel Dis 2020; 26:1068-1076. [PMID: 31587061 DOI: 10.1093/ibd/izz225] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND The incidence of pediatric inflammatory bowel diseases (PIBDs: Crohn's disease [CD], ulcerative colitis [UC]) is on the rise around the world. Yet, the critical risk factors for this rising incidence are not well understood. Demographic characteristics of PIBD may improve our understanding of their developmental origins and aid in prevention. METHODS Four hundred eighty-eight consecutive PIBD patients diagnosed at Texas Children's Hospital from 13 counties around Houston were studied. An annual incidence map was created by ZIP code of residence at diagnosis by using ArcGIS and the American Community Survey from the US Census Bureau. Correlation between demographic variables and PIBD incidence was examined. A model to explain incidence from different health factors was created in R. RESULTS Hispanic children were more likely to be diagnosed with UC (P < 0.01) and unclassified IBD (IBD-U) (P < 0.03) compared with other races/ethnicities. A significant positive correlation (r = 0.35, P < 0.0001) between median household income and PIBD incidence was observed (UC: r = 0.23, P < 0.0001; CD: r = 0.22, P = 0.0004). ZIP codes with majority college-educated adults had a higher incidence of PIBD than ZIP codes with majority high school-educated adults (P < 0.0001). Pediatric cases with CD were more common in ZIP codes where the majority of adults were college educated (P < 0.0001). Pediatric cases with UC, however, were more common in ZIP codes where the majority of adults were high school educated (P = 0.0036). CONCLUSIONS Hispanic children more commonly present with UC and IBD-U in southern USA. Household income and/or adult education-related environmental/dietary differences may be important in the developmental origins of PIBD in large metro areas, such as Houston.
Collapse
Affiliation(s)
- Mahesh Krishna
- Wiess School of Natural Sciences, Rice University, Houston, TX, USA.,Section of Pediatric Gastroenterology, Baylor College of Medicine, Houston, TX, USA
| | - Aziza Salako
- Wiess School of Natural Sciences, Rice University, Houston, TX, USA
| | - Tatiana Fofanova
- Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Richard Kellermayer
- Section of Pediatric Gastroenterology, Baylor College of Medicine, Houston, TX, USA.,USDA/ARS Children's Nutrition Research Center, Texas Children's Hospital, Houston, TX, USA
| |
Collapse
|
6
|
Britto S, Kellermayer R. Carbohydrate Monotony as Protection and Treatment for Inflammatory Bowel Disease. J Crohns Colitis 2019; 13:942-948. [PMID: 30715243 DOI: 10.1093/ecco-jcc/jjz011] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 12/17/2018] [Accepted: 01/24/2019] [Indexed: 02/07/2023]
Abstract
The nutritional developmental origins of inflammatory bowel disease[s] (IBDs: Crohn's disease or Crohn disease [CD] and ulcerative colitis [UC]) and their diet-based treatments continue to receive increasing attention. There is growing evidence for the success of nutrition-based treatments, such as exclusive enteral nutrition [EEN] and the specific carbohydrate diet [SCD], in both paediatric and adult patients. Beyond these two dietary interventions, symptomatic benefit in IBD has also been shown from a gluten-free diet [GFD] and paleolithic diet [PALEO], among others. These nutritional therapies may point to critical factors in not only the pathology, but also the pathogenesis of the disease group. It is difficult, however, to discern a common element within the large number of diet-based causation theories [e.g. emulsifiers, processed foods, refrigeration, increased total fat intake, low fibre intake, carbohydrate dominant food, etc.] and the varied dietary treatments of IBD. This Viewpoint article highlights that carbohydrate variation links diet-based causation theories, and that carbohydrate monotony or persistence is the commonly shared characteristic of diet-based IBD therapy. Further research directed towards carbohydrate monotony may critically advance the prevention and treatment of these highly morbid conditions.
Collapse
Affiliation(s)
- Savini Britto
- Section of Pediatric Gastroenterology, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
| | - Richard Kellermayer
- Section of Pediatric Gastroenterology, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA.,USDA/ARS Children's Nutrition Research Center, Houston, TX, USA
| |
Collapse
|
7
|
Castro F, de Souza HSP. Dietary Composition and Effects in Inflammatory Bowel Disease. Nutrients 2019; 11:1398. [PMID: 31234325 PMCID: PMC6628370 DOI: 10.3390/nu11061398] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Revised: 06/14/2019] [Accepted: 06/19/2019] [Indexed: 02/07/2023] Open
Abstract
Dramatic changes in the environment and human lifestyle have been associated with the rise of various chronic complex diseases, such as inflammatory bowel disease (IBD). A dysbiotic gut microbiota has been proposed as a crucial pathogenic element, contributing to immune imbalances and fostering a proinflammatory milieu, which may be associated with disease relapses or even the initiation of IBD. In addition to representing important regulators of the mucosal immunity and the composition of the gut microbiota, food components have been shown to be potential environmental triggers of epigenetic modifications. In the context of chronic intestinal inflammation, dietary habits and specific food components have been implicated as important modulators of epigenetic mechanisms, including DNA methylation, which may predispose a person to the increased risk of the initiation and evolution of IBD. This review provides novel insights about how dietary factors may interact with the intestinal mucosa and modulate immune homeostasis by shaping the intestinal ecosystem, as well as the potential influence of diet in the etiopathogenesis and management of IBD.
Collapse
Affiliation(s)
- Fernando Castro
- Department of Internal Medicine, School of Medicine, Federal University of Rio de Janeiro, 21941-913 Rio de Janeiro, Brazil.
| | - Heitor S P de Souza
- Department of Internal Medicine, School of Medicine, Federal University of Rio de Janeiro, 21941-913 Rio de Janeiro, Brazil.
- Department of Internal Medicine, D'Or Institute for Research and Education (IDOR), 22281-100 Rio de Janeiro, Brazil.
| |
Collapse
|
8
|
Ihekweazu FD, Fofanova TY, Queliza K, Nagy-Szakal D, Stewart CJ, Engevik MA, Hulten KG, Tatevian N, Graham DY, Versalovic J, Petrosino JF, Kellermayer R. Bacteroides ovatus ATCC 8483 monotherapy is superior to traditional fecal transplant and multi-strain bacteriotherapy in a murine colitis model. Gut Microbes 2019; 10:504-520. [PMID: 30663928 PMCID: PMC6748610 DOI: 10.1080/19490976.2018.1560753] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Background and aims: Bacteriotherapy aimed at addressing dysbiosis may be therapeutic for Inflammatory Bowel Diseases (IBDs). We sought to determine if defined Bacteroides-based bacteriotherapy could be an effective and consistent alternative to fecal microbiota transplantation (FMT) in a murine model of IBD. Methods: We induced experimental colitis in 8- 12-week-old C57BL/6 mice using 2-3% dextran sodium sulfate. Mice were simultaneously treated by oral gavage with a triple-Bacteroides cocktail, individual Bacteroides strains, FMT using stool from healthy donor mice, or their own stool as a control. Survival, weight loss and markers of inflammation (histology, serum amyloid A, cytokine production) were correlated to 16S rRNA gene profiling of fecal and mucosal microbiomes. Results: Triple-Bacteroides combination therapy was more protective against weight loss and mortality than traditional FMT therapy. B. ovatus ATCC8483 was more effective than any individual strain, or a combination of strains, in preventing weight loss, decreasing histological damage, dampening inflammatory response, and stimulating epithelial recovery. Irrespective of the treatment group, overall Bacteroides abundance associated with treatment success and decreased cytokine production while the presence of Akkermansia correlated with treatment failure. However, the therapeutic benefit associated with high Bacteroides abundance was negated in the presence of Streptococcus. Conclusions: Bacteroides ovatus monotherapy was more consistent and effective than traditional FMT at ameliorating colitis and stimulating epithelial recovery in a murine model of IBD. Given the tolerability of Bacteroides ovatus ATCC 8483 in an active, on-going human study, this therapy may be repurposed for the management of IBD in a clinically expedient timeline.
Collapse
Affiliation(s)
- Faith D. Ihekweazu
- Pediatric Gastroenterology, Hepatology and Nutrition, Baylor College of Medicine, Texas Children’s Hospital, Houston, TX, USA,CONTACT Faith D. Ihekweazu Pediatric Gastroenterology, Hepatology and Nutrition, Baylor College of Medicine, Texas Children’s Hospital, 1102 Bates Ave, FT860.28, Houston, TX 77030, USA
| | | | - Karen Queliza
- Pediatric Gastroenterology, Hepatology and Nutrition, Baylor College of Medicine, Texas Children’s Hospital, Houston, TX, USA
| | - Dorottya Nagy-Szakal
- Center for Infection and Immunity, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Christopher J. Stewart
- Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA,Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Melinda A. Engevik
- Pediatric Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Kristina G. Hulten
- Pediatric Infectious Disease, Baylor College of Medicine, Texas Children’s Hospital, Houston, TX, USA
| | - Nina Tatevian
- Pathology and Laboratory Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - David Y. Graham
- Gastroenterology, Baylor College of Medicine, Houston, TX, USA
| | - James Versalovic
- Pediatric Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
| | | | - Richard Kellermayer
- Pediatric Gastroenterology, Hepatology and Nutrition, Baylor College of Medicine, Texas Children’s Hospital, Houston, TX, USA
| |
Collapse
|
9
|
Korte SW, Franklin CL, Dorfmeyer RA, Ericsson AC. Effects of Fenbendazole-impregnated Feed and Topical Moxidectin during Quarantine on the Gut Microbiota of C57BL/6 Mice. JOURNAL OF THE AMERICAN ASSOCIATION FOR LABORATORY ANIMAL SCIENCE : JAALAS 2018; 57:229-235. [PMID: 29784074 PMCID: PMC5966229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 12/09/2017] [Accepted: 12/13/2017] [Indexed: 06/08/2023]
Abstract
To protect the biosecurity of research rodent colonies, research institutions frequently require a quarantine period for live animals transferred into their facilities. Quarantine practices often include antibiotic and antiparasitic treatment with drugs such as fenbendazole and macrolide lactones. The influence of these compounds on the resident gut microbiota of mice is unknown, and any effects might subsequently affect model reproducibility. To test the influence of standard quarantine procedures on the composition of the microbiota, C57BL/6 mice, purchased from 2 different commercial suppliers, were randomly assigned to treatment groups (n = 12) by vendor and treated with fenbendazole-supplemented feed, topical moxidectin, both treatments, or no treatment (control), according to our institution's standard treatment regimen and duration. Feces were collected on arrival, immediately after completing the 8-wk treatment, and at 2 and 4 wk after treatment. Fecal DNA was extracted, sequenced, and analyzed to compare the changes in the microbiota of treated and control groups. Although significant main effects of time and treatment and interactions between those variables were detected in comparisons of richness, α-diversity, and β-diversity, the effect sizes associated with any particular treatment were consistently much smaller than that associated with acclimation to a new facility in the absence of any quarantine treatments. This outcome, along with the visual evaluation of principal coordinate analysis based on multiple similarity indices, suggests that time or institution plays a larger role in alterations of the murine gut microbiota than do quarantine treatments on its composition.
Collapse
Affiliation(s)
- Scott W Korte
- Office of Animal Resources, University of Missouri, Columbia, Missouri
| | - Craig L Franklin
- Metagenomics Center, Mutant Mouse Resource and Research Center, Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri
| | - Rebecca A Dorfmeyer
- Mutant Mouse Resource and Research Center, University of Missouri, Columbia, Missouri
| | - Aaron C Ericsson
- Metagenomics Center, Mutant Mouse Resource and Research Center, Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri;,
| |
Collapse
|
10
|
Zhang R, Ma YP, Liu WT. Effect of maternal methyl donor deficient diet on experimental colitis in rat offsprings. Shijie Huaren Xiaohua Zazhi 2017; 25:2791-2797. [DOI: 10.11569/wcjd.v25.i31.2791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate whether maternal methyl donor deficiency promotes the pathogenesis and development of ulcerative colitis (UC) by affecting DNA methylation.
METHODS Maternal BALB/c mice were fed either a normal diet (C) or a methyl donor deficiency diet (D) from one month before pregnancy until the offspring weaned. Young mice were given dextran sulfate sodium (DSS) or purified water as drink water for five days. The young mice were randomly divided into four groups: C/DSS-, D/DSS-, C/DSS+, and D/DSS+. The degree of colonic inflammation was evaluated; serum folate, vitamin B12, and homocysteine (Hcy) were determined; the expression of interferon-γ (IFN-γ) in the colonic mucosa was detected; and the methylation level of CpG islands in the IFNG promoter was determined.
RESULTS The disease activity index (DAI) was significantly higher in the D/DSS+ group than in the C/DSS+ group (3.22 ± 0.55 vs 2.22 ± 0.50, P < 0.01). Compared to the normal diet group, the methyl donor deficient diet group had significantly lower serum levels of folic acid (8.87 nmol/L ± 1.11 nmol/L vs 11.34 nmol/L ± 0.31 nmol/L, P < 0.01) and vitamin B12 (409.2 ng/L ± 56.27 ng/L vs 676.1 ng/L ± 1.66 ng/L, P < 0.01) and significantly higher Hcy (8.45 μmol/L ± 0.35 μmol/L vs 6.77 μmol/L ± 0.36 μmol/L, P < 0.01). The IFN-γ expression in the colonic mucosa was significantly higher in the C/DSS+ and D/DSS+ groups than in the C/DSS- and D/DSS- groups (5.3 ± 1.2 vs 10.6 ± 10.8, χ2 = 14.517, P < 0.01). The expression level of IFN-γ in the colonic mucosa of DSS+ groups was positively related to the degree of inflammation (r = 0.853, 0.840; P = 0.031, 0.036; P < 0.05). There were no significant differences in the methylation level of all CpG sites or same CpG sites among the four samples.
CONCLUSION Methyl donor deficient diet may cause aggravated experimental colitis by increasing the expression of IFN-γ. The abnormal expression of IFN-γ in the colonic mucosa has no relationship with hypo-methylation of CpG islands in the IFNG promoter.
Collapse
Affiliation(s)
- Rui Zhang
- Department of Gastroenterology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Yu-Ping Ma
- Department of Gastroenterology, Xingtai People's Hospital, Xingtai 054000, Hebei Province, China
| | - Wen-Tian Liu
- Department of Gastroenterology, Tianjin Medical University General Hospital, Tianjin 300052, China
| |
Collapse
|
11
|
Cuív PÓ, Begun J, Keely S, Lewindon PJ, Morrison M. Towards an integrated understanding of the therapeutic utility of exclusive enteral nutrition in the treatment of Crohn's disease. Food Funct 2017; 7:1741-51. [PMID: 26948398 DOI: 10.1039/c5fo01196e] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Crohn's disease (CD) is a chronic disease characterized by episodic and disabling inflammation of the gastrointestinal tract in genetically susceptible individuals. The incidence and prevalence of CD is rising rapidly across the world emphasising that disease risk is also influenced by environmental and lifestyle factors, as well as the microbial community resident in the gut. Childhood-onset CD is associated with an aggressive disease course that can adversely impact patient growth and development. There is no cure for CD however new onset and recurrent cases of paediatric CD are often responsive to exclusive enteral nutrition (EEN) treatment. EEN treatment involves the exclusive consumption of an elemental or polymeric formula for several weeks and it is well established as a primary intervention strategy. EEN treatments typically achieve remission rates of over 80% and importantly they are associated with a high rate of mucosal healing, far superior to steroids, which is prognostic of improved long-term health outcomes. Furthermore, they are safe, have few side effects, and improve nutritional status and linear growth. Surprisingly, despite the utility of EEN our understanding of the host-microbe-diet interactions that underpin clinical remission and mucosal healing are limited. Here, we review the current state of knowledge and propose that the induction of autophagy, in addition to modulation of the microbiota and coordinated effects on inflammation and epithelial cell biology, may be critical for the therapeutic effects associated with EEN. A better understanding of EEN treatment will provide new opportunities to restore gut homeostasis and prolong periods of remission, as well as provide new insights into the factors that trigger and perhaps prevent CD.
Collapse
Affiliation(s)
- Páraic Ó Cuív
- The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, QLD 4102, Australia.
| | - Jakob Begun
- Mater Research Institute, The University of Queensland, Brisbane, QLD 4102, Australia and School of Medicine, The University of Queensland, Brisbane, QLD 4102, Australia and Department of Gastroenterology, Mater Health Services, Brisbane, Australia
| | - Simon Keely
- School of Biomedical Science and Pharmacy, The University of Newcastle, University Drive, Callaghan, NSW 2308, Australia and Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
| | - Peter J Lewindon
- The University of Queensland, Department of Paediatrics and Child Health, Lady Cilento Children's Hospital, QLD 4101, Australia
| | - Mark Morrison
- The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, QLD 4102, Australia.
| |
Collapse
|
12
|
Microbiome-Epigenome Interactions and the Environmental Origins of Inflammatory Bowel Diseases. J Pediatr Gastroenterol Nutr 2016; 62:208-19. [PMID: 26308318 PMCID: PMC4724338 DOI: 10.1097/mpg.0000000000000950] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The incidence of pediatric inflammatory bowel disease (IBD), which includes Crohn disease and ulcerative colitis, has risen alarmingly in the Western and developing world in recent decades. Epidemiologic (including monozygotic twin and migrant) studies highlight the substantial role of environment and nutrition in IBD etiology. Here we review the literature supporting the developmental and environmental origins hypothesis of IBD. We also provide a detailed exploration of how the human microbiome and epigenome (primarily through DNA methylation) may be important elements in the developmental origins of IBD in both children and adults.
Collapse
|
13
|
Lee D, Albenberg L, Compher C, Baldassano R, Piccoli D, Lewis JD, Wu GD. Diet in the pathogenesis and treatment of inflammatory bowel diseases. Gastroenterology 2015; 148:1087-106. [PMID: 25597840 PMCID: PMC4409494 DOI: 10.1053/j.gastro.2015.01.007] [Citation(s) in RCA: 284] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Revised: 01/12/2015] [Accepted: 01/13/2015] [Indexed: 12/14/2022]
Abstract
Some of the most common symptoms of the inflammatory bowel diseases (IBD, which include ulcerative colitis and Crohn's disease) are abdominal pain, diarrhea, and weight loss. It is therefore not surprising that clinicians and patients have wondered whether dietary patterns influence the onset or course of IBD. The question of what to eat is among the most commonly asked by patients, and among the most difficult to answer for clinicians. There are substantial variations in dietary behaviors of patients and recommendations for them, although clinicians do not routinely endorse specific diets for patients with IBD. Dietary clinical trials have been limited by their inability to include a placebo control, contamination of study groups, and inclusion of patients receiving medical therapies. Additional challenges include accuracy of information on dietary intake, complex interactions between foods consumed, and differences in food metabolism among individuals. We review the roles of diet in the etiology and management of IBD based on plausible mechanisms and clinical evidence. Researchers have learned much about the effects of diet on the mucosal immune system, epithelial function, and the intestinal microbiome; these findings could have significant practical implications. Controlled studies of patients receiving enteral nutrition and observations made from patients on exclusion diets have shown that components of whole foods can have deleterious effects for patients with IBD. Additionally, studies in animal models suggested that certain nutrients can reduce intestinal inflammation. In the future, engineered diets that restrict deleterious components but supplement beneficial nutrients could be used to modify the luminal intestinal environment of patients with IBD; these might be used alone or in combination with immunosuppressive agents, or as salvage therapy for patients who do not respond or lose responsiveness to medical therapies. Stricter diets might be required to induce remission, and more sustainable exclusion diets could be used to maintain long-term remission.
Collapse
Affiliation(s)
| | | | | | | | | | - James D. Lewis
- Co-Corresponding authors: James D. Lewis, Professor of Medicine and Epidemiology, Division of Gastroenterology, Perelman School of Medicine, University of Pennsylvania, 720 Blockley Hall, 423 Guardian Drive, Philadelphia, PA 19104-6021, Office: (215) 573-5137, Fax: (215) 573-0813, ; Gary D. Wu, Professor of Medicine, Division of Gastroenterology, Perelman School of Medicine, University of Pennsylvania, Suite 915, Biomedical Research Building, 421 Curie Boulevard, Philadelphia, PA 19104, Office: (215) 898-0158, Fax: (215) 573-2024,
| | - Gary D. Wu
- Co-Corresponding authors: James D. Lewis, Professor of Medicine and Epidemiology, Division of Gastroenterology, Perelman School of Medicine, University of Pennsylvania, 720 Blockley Hall, 423 Guardian Drive, Philadelphia, PA 19104-6021, Office: (215) 573-5137, Fax: (215) 573-0813, ; Gary D. Wu, Professor of Medicine, Division of Gastroenterology, Perelman School of Medicine, University of Pennsylvania, Suite 915, Biomedical Research Building, 421 Curie Boulevard, Philadelphia, PA 19104, Office: (215) 898-0158, Fax: (215) 573-2024,
| |
Collapse
|
14
|
Nagy-Szakal D, Mir SAV, Harris RA, Dowd SE, Yamada T, Lacorazza HD, Tatevian N, Smith CW, de Zoeten EF, Klein J, Kellermayer R. Loss of n-6 fatty acid induced pediatric obesity protects against acute murine colitis. FASEB J 2015; 29:3151-9. [PMID: 25903104 DOI: 10.1096/fj.14-267690] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 03/31/2015] [Indexed: 12/22/2022]
Abstract
Dietary influences may affect microbiome composition and host immune responses, thereby modulating propensity toward inflammatory bowel diseases (IBDs): Crohn disease (CD) and ulcerative colitis (UC). Dietary n-6 fatty acids have been associated with UC in prospective studies. However, the critical developmental period when (n-6) consumption may induce UC is not known. We examined the effects of transiently increased n-6 consumption during pediatric development on subsequent dextran-sulfate-sodium (DSS)-induced acute murine colitis. The animals transiently became obese then rapidly lost this phenotype. Interestingly, mice were protected against DSS colitis 40 days after n-6 consumption. The transient high n-6-induced protection against colitis was fat type- and dietary reversal-dependent and could be transferred to germ-free mice by fecal microbiota transplantation. We also detected decreased numbers of chemokine receptor (Cxcr)5(+) CD4(+) T cells in the mesenteric lymph nodes (MLNs) of transiently n-6-fed mice. Further experiments revealed that anti-chemokine ligand (Cxcl)13 (the ligand of Cxcr5) antibody treatment decreased DSS colitis severity, implicating the importance of the Cxcr5-Cxcl13 pathway in mammalian colitis. Consecutively, we found elevated CXCL13 concentrations (CD: 1.8-fold, P = 0.0077; UC: 1.9-fold, P = 0.056) in the serum of untreated pediatric IBD patients. The human serologic observations supported the translational relevance of our findings.
Collapse
Affiliation(s)
- Dorottya Nagy-Szakal
- *Section of Pediatric Gastroenterology, Baylor College of Medicine, Texas Children Hospital, Houston, Texas, USA; U.S. Department of Agriculture/Agricultural Research Service Children's Nutrition Research Center, Houston, Texas, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA; Molecular Research LP, Shallowater, Texas, USA; Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, USA; Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center, Houston, Texas, USA; Digestive Health Institute, Children's Hospital Colorado, Aurora, Colorado, USA; and **School of Dentistry, The University of Texas Health Science Center, Houston, Texas, USA
| | - Sabina A V Mir
- *Section of Pediatric Gastroenterology, Baylor College of Medicine, Texas Children Hospital, Houston, Texas, USA; U.S. Department of Agriculture/Agricultural Research Service Children's Nutrition Research Center, Houston, Texas, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA; Molecular Research LP, Shallowater, Texas, USA; Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, USA; Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center, Houston, Texas, USA; Digestive Health Institute, Children's Hospital Colorado, Aurora, Colorado, USA; and **School of Dentistry, The University of Texas Health Science Center, Houston, Texas, USA
| | - R Alan Harris
- *Section of Pediatric Gastroenterology, Baylor College of Medicine, Texas Children Hospital, Houston, Texas, USA; U.S. Department of Agriculture/Agricultural Research Service Children's Nutrition Research Center, Houston, Texas, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA; Molecular Research LP, Shallowater, Texas, USA; Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, USA; Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center, Houston, Texas, USA; Digestive Health Institute, Children's Hospital Colorado, Aurora, Colorado, USA; and **School of Dentistry, The University of Texas Health Science Center, Houston, Texas, USA
| | - Scot E Dowd
- *Section of Pediatric Gastroenterology, Baylor College of Medicine, Texas Children Hospital, Houston, Texas, USA; U.S. Department of Agriculture/Agricultural Research Service Children's Nutrition Research Center, Houston, Texas, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA; Molecular Research LP, Shallowater, Texas, USA; Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, USA; Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center, Houston, Texas, USA; Digestive Health Institute, Children's Hospital Colorado, Aurora, Colorado, USA; and **School of Dentistry, The University of Texas Health Science Center, Houston, Texas, USA
| | - Takeshi Yamada
- *Section of Pediatric Gastroenterology, Baylor College of Medicine, Texas Children Hospital, Houston, Texas, USA; U.S. Department of Agriculture/Agricultural Research Service Children's Nutrition Research Center, Houston, Texas, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA; Molecular Research LP, Shallowater, Texas, USA; Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, USA; Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center, Houston, Texas, USA; Digestive Health Institute, Children's Hospital Colorado, Aurora, Colorado, USA; and **School of Dentistry, The University of Texas Health Science Center, Houston, Texas, USA
| | - H Daniel Lacorazza
- *Section of Pediatric Gastroenterology, Baylor College of Medicine, Texas Children Hospital, Houston, Texas, USA; U.S. Department of Agriculture/Agricultural Research Service Children's Nutrition Research Center, Houston, Texas, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA; Molecular Research LP, Shallowater, Texas, USA; Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, USA; Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center, Houston, Texas, USA; Digestive Health Institute, Children's Hospital Colorado, Aurora, Colorado, USA; and **School of Dentistry, The University of Texas Health Science Center, Houston, Texas, USA
| | - Nina Tatevian
- *Section of Pediatric Gastroenterology, Baylor College of Medicine, Texas Children Hospital, Houston, Texas, USA; U.S. Department of Agriculture/Agricultural Research Service Children's Nutrition Research Center, Houston, Texas, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA; Molecular Research LP, Shallowater, Texas, USA; Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, USA; Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center, Houston, Texas, USA; Digestive Health Institute, Children's Hospital Colorado, Aurora, Colorado, USA; and **School of Dentistry, The University of Texas Health Science Center, Houston, Texas, USA
| | - C Wayne Smith
- *Section of Pediatric Gastroenterology, Baylor College of Medicine, Texas Children Hospital, Houston, Texas, USA; U.S. Department of Agriculture/Agricultural Research Service Children's Nutrition Research Center, Houston, Texas, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA; Molecular Research LP, Shallowater, Texas, USA; Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, USA; Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center, Houston, Texas, USA; Digestive Health Institute, Children's Hospital Colorado, Aurora, Colorado, USA; and **School of Dentistry, The University of Texas Health Science Center, Houston, Texas, USA
| | - Edwin F de Zoeten
- *Section of Pediatric Gastroenterology, Baylor College of Medicine, Texas Children Hospital, Houston, Texas, USA; U.S. Department of Agriculture/Agricultural Research Service Children's Nutrition Research Center, Houston, Texas, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA; Molecular Research LP, Shallowater, Texas, USA; Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, USA; Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center, Houston, Texas, USA; Digestive Health Institute, Children's Hospital Colorado, Aurora, Colorado, USA; and **School of Dentistry, The University of Texas Health Science Center, Houston, Texas, USA
| | - John Klein
- *Section of Pediatric Gastroenterology, Baylor College of Medicine, Texas Children Hospital, Houston, Texas, USA; U.S. Department of Agriculture/Agricultural Research Service Children's Nutrition Research Center, Houston, Texas, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA; Molecular Research LP, Shallowater, Texas, USA; Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, USA; Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center, Houston, Texas, USA; Digestive Health Institute, Children's Hospital Colorado, Aurora, Colorado, USA; and **School of Dentistry, The University of Texas Health Science Center, Houston, Texas, USA
| | - Richard Kellermayer
- *Section of Pediatric Gastroenterology, Baylor College of Medicine, Texas Children Hospital, Houston, Texas, USA; U.S. Department of Agriculture/Agricultural Research Service Children's Nutrition Research Center, Houston, Texas, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA; Molecular Research LP, Shallowater, Texas, USA; Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, USA; Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center, Houston, Texas, USA; Digestive Health Institute, Children's Hospital Colorado, Aurora, Colorado, USA; and **School of Dentistry, The University of Texas Health Science Center, Houston, Texas, USA
| |
Collapse
|
15
|
Shah R, Kellermayer R. Microbiome associations of therapeutic enteral nutrition. Nutrients 2014; 6:5298-311. [PMID: 25421531 PMCID: PMC4245590 DOI: 10.3390/nu6115298] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Revised: 10/08/2014] [Accepted: 10/21/2014] [Indexed: 12/19/2022] Open
Abstract
One of the most effective forms of therapeutic enteral nutrition is designated as “exclusive enteral nutrition” (EEN). EEN constitutes the monotonous enteral delivery of complete liquid nutrition and has been most explored in the treatment Crohn’s disease (CD), a form of inflammatory bowel disease. While EEN’s mechanisms of action are not clearly understood, it has been shown to modify the composition of the intestinal microbiome, an important component of CD pathogenesis. The current literature on the intestinal microbiome in healthy individuals and CD patients is reviewed with respect to EEN therapy. Further investigations in this field are needed to better understand the role and potential for EEN in chronic human disorders.
Collapse
Affiliation(s)
- Rajesh Shah
- Department of Internal Medicine, Section of Gastroenterology, Baylor College of Medicine, Houston, TX, 77030, USA.
| | - Richard Kellermayer
- Department of Pediatrics, Section of Gastroenterology, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, 77030, USA.
| |
Collapse
|
16
|
Fleet JC. Animal models of gastrointestinal and liver diseases. New mouse models for studying dietary prevention of colorectal cancer. Am J Physiol Gastrointest Liver Physiol 2014; 307:G249-59. [PMID: 24875098 PMCID: PMC4121636 DOI: 10.1152/ajpgi.00019.2014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Colorectal cancer is a heterogeneous disease that is one of the major causes of cancer death in the U.S. There is evidence that lifestyle factors like diet can modulate the course of this disease. Demonstrating the benefit and mechanism of action of dietary interventions against colon cancer will require studies in preclinical models. Many mouse models have been developed to study colon cancer but no single model can reflect all types of colon cancer in terms of molecular etiology. In addition, many models develop only low-grade cancers and are confounded by development of the disease outside of the colon. This review will discuss how mice can be used to model human colon cancer and it will describe a variety of new mouse models that develop colon-restricted cancer as well as more advanced phenotypes for studies of late-state disease.
Collapse
Affiliation(s)
- James C. Fleet
- 1Department of Nutrition Science, Purdue University, West Lafayette, Indiana; and ,2Purdue University Center for Cancer Research, Purdue University, West Lafayette, Indiana
| |
Collapse
|
17
|
Arató A. Milestones in understanding of the pathogenesis of immunmediated intestinal disorders. Development of their diagnosis and therapy. Orv Hetil 2013; 154:1512-23. [DOI: 10.1556/oh.2013.29710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
In the last decades our knowledge has been enormously broadened about the structure and function of the gut associated lymphoid system. It was recognized how intricate and finely tuned connection exists between the gut bacterial flora and the intestinal mucosa. This subtle balance ensures mucosal homeostasis, which has a key role in organ defence against pathogens. However, at the same time this system makes possible the development of oral tolerance toward the commensals and the food antigens. In case of any disturbances in this finely tuned process, immunmediated intestinal disorders may easily develop. The first part of this paper reviews the structure and function of the mucosal immune system, while the second part surveys the pathogenesis, diagnosis and therapy of coeliac disease, inflammatory bowel disease and cow’s milk allergy induced enteropathy. Orv. Hetil., 2013, 154, 1512–1523.
Collapse
Affiliation(s)
- András Arató
- Semmelweis Egyetem, Általános Orvostudományi Kar I. Gyermekgyógyászati Klinika Budapest Bókay János u. 53. 1083
| |
Collapse
|
18
|
Mir SA, Nagy-Szakal D, Dowd SE, Szigeti RG, Smith CW, Kellermayer R. Prenatal methyl-donor supplementation augments colitis in young adult mice. PLoS One 2013; 8:e73162. [PMID: 23977377 PMCID: PMC3747105 DOI: 10.1371/journal.pone.0073162] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Accepted: 07/17/2013] [Indexed: 12/17/2022] Open
Abstract
Inflammatory bowel diseases (IBD) have become highly prevalent in developed countries. Environmentally triggered exaggerated immune responses against the intestinal microbiome are thought to mediate the disorders. The potential dietary origins of the disease group have been implicated. However, the effects of environmental influences on prenatal developmental programming in respect to orchestrating postnatal microbiome composition and predilection towards mammalian colitis have not been examined. We tested how transient prenatal exposure to methyl donor micronutrient (MD) supplemented diets may impact predilection towards IBD in a murine dextran sulfate sodium (DSS) colitis model. Prenatal MD supplementation was sufficient to modulate colonic mucosal Ppara expression (3.2 fold increase; p=0.022) and worsen DSS colitis in young adulthood. The prenatal dietary exposure shifted the postnatal colonic mucosal and cecal content microbiomes. Transfer of the gut microbiome from prenatally MD supplemented young adult animals into germ free mice resulted in increased colitis susceptibility in the recipients compared to controls. Therefore, the prenatal dietary intervention induced the postnatal nurturing of a colitogenic microbiome. Our results show that prenatal nutritional programming can modulate the mammalian host to harbor a colitogenic microbiome. These findings may be relevant for the nutritional developmental origins of IBD.
Collapse
Affiliation(s)
- Sabina A. Mir
- Section of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Baylor College of Medicine, Texas Children’s Hospital, Department of Agriculture/ARS Children’s Nutrition Research Center, Houston, Texas, United States of America
| | - Dorottya Nagy-Szakal
- Section of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Baylor College of Medicine, Texas Children’s Hospital, Department of Agriculture/ARS Children’s Nutrition Research Center, Houston, Texas, United States of America
| | - Scot E. Dowd
- MR DNA (Molecular Research), Shallowater, Texas, United States of America
| | - Reka G. Szigeti
- Department of Pathology, Baylor College of Medicine, Houston, Texas, United States of America
| | - C. Wayne Smith
- Section of Infectious Disease, Department of Pediatrics, Baylor College of Medicine, Department of Agriculture/ARS Children’s Nutrition Research Center, Houston, Texas, United States of America
| | - Richard Kellermayer
- Section of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Baylor College of Medicine, Texas Children’s Hospital, Department of Agriculture/ARS Children’s Nutrition Research Center, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
19
|
Nagy-Szakal D, Hollister EB, Luna RA, Szigeti R, Tatevian N, Smith CW, Versalovic J, Kellermayer R. Cellulose supplementation early in life ameliorates colitis in adult mice. PLoS One 2013; 8:e56685. [PMID: 23437211 PMCID: PMC3577696 DOI: 10.1371/journal.pone.0056685] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2012] [Accepted: 01/14/2013] [Indexed: 01/19/2023] Open
Abstract
Decreased consumption of dietary fibers, such as cellulose, has been proposed to promote the emergence of inflammatory bowel diseases (IBD: Crohn disease [CD] and ulcerative colitis [UC]) where intestinal microbes are recognized to play an etiologic role. However, it is not known if transient fiber consumption during critical developmental periods may prevent consecutive intestinal inflammation. The incidence of IBD peaks in young adulthood indicating that pediatric environmental exposures may be important in the etiology of this disease group. We studied the effects of transient dietary cellulose supplementation on dextran sulfate sodium (DSS) colitis susceptibility during the pediatric period in mice. Cellulose supplementation stimulated substantial shifts in the colonic mucosal microbiome. Several bacterial taxa decreased in relative abundance (e.g., Coriobacteriaceae [p = 0.001]), and other taxa increased in abundance (e.g., Peptostreptococcaceae [p = 0.008] and Clostridiaceae [p = 0.048]). Some of these shifts persisted for 10 days following the cessation of cellulose supplementation. The changes in the gut microbiome were associated with transient trophic and anticolitic effects 10 days following the cessation of a cellulose-enriched diet, but these changes diminished by 40 days following reversal to a low cellulose diet. These findings emphasize the transient protective effect of dietary cellulose in the mammalian large bowel and highlight the potential role of dietary fibers in amelioration of intestinal inflammation.
Collapse
Affiliation(s)
- Dorottya Nagy-Szakal
- Section of Pediatric Gastroenterology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States of America
- USDA/ARS Children's Nutrition Research Center, Houston, Texas, United States of America
| | - Emily B. Hollister
- Department of Pathology, Baylor College of Medicine, Houston, Texas, United States of America
- Texas Children's Hospital, Houston, Texas, United States of America
| | - Ruth Ann Luna
- Department of Pathology, Baylor College of Medicine, Houston, Texas, United States of America
- Texas Children's Hospital, Houston, Texas, United States of America
| | - Reka Szigeti
- Department of Pathology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Nina Tatevian
- Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center, Houston, Texas, United States of America
| | - C. Wayne Smith
- USDA/ARS Children's Nutrition Research Center, Houston, Texas, United States of America
- Texas Children's Hospital, Houston, Texas, United States of America
| | - James Versalovic
- Department of Pathology, Baylor College of Medicine, Houston, Texas, United States of America
- Texas Children's Hospital, Houston, Texas, United States of America
| | - Richard Kellermayer
- Section of Pediatric Gastroenterology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States of America
- USDA/ARS Children's Nutrition Research Center, Houston, Texas, United States of America
- Texas Children's Hospital, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|