51
|
Rahman S, Davids M, van Hamersveld PHP, Welting O, Rahaoui H, Schuren F, Meijer SL, van den Wijngaard RM, Hakvoort TBM, de Jonge WJ, Heinsbroek SEM. Dietary Curdlan Enhances Bifidobacteria and Reduces Intestinal Inflammation in Mice. Nutrients 2021; 13:1305. [PMID: 33920960 PMCID: PMC8071228 DOI: 10.3390/nu13041305] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 04/12/2021] [Indexed: 12/12/2022] Open
Abstract
β-glucan consumption is known for its beneficial health effects, but the mode of action is unclear. While humans and mice lack the required enzymes to digest β-glucans, certain intestinal microbes can digest β-glucans, triggering gut microbial changes. Curdlan, a particulate β-glucan isolated from Alcaligenes faecalis, is used as a food additive. In this study we determined the effect of curdlan intake in mice on the intestinal microbiota and dextran sodium sulfate (DSS)-induced intestinal inflammation. The effect of curdlan on the human intestinal microbiota was assessed using i-screen, an assay for studying anaerobic microbial interactions. Mice received oral gavage with vehicle or curdlan for 14 days followed by DSS for 7 days. The curdlan-fed group showed reduced weight loss and colonic inflammation compared to the vehicle-fed group. Curdlan intake did not induce general microbiota community changes, although a specific Bifidobacterium, closely related to Bifidobacterium choerinum, was observed to be 10- to 100-fold more prevalent in the curdlan-fed group under control and colitis conditions, respectively. When tested in i-screen, curdlan induced a global change in the microbial composition of the healthy intestinal microbiota from a human. Overall, these results suggest that dietary curdlan induces microbiota changes that could reduce intestinal inflammation.
Collapse
Affiliation(s)
- Shafaque Rahman
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology and Metabolism, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, 1105 BK Amsterdam, The Netherlands; (S.R.); (P.H.P.v.H.); (O.W.); (R.M.v.d.W.); (T.B.M.H.); (W.J.d.J.)
| | - Mark Davids
- Department of Vascular Medicine, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands;
| | - Patricia H. P. van Hamersveld
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology and Metabolism, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, 1105 BK Amsterdam, The Netherlands; (S.R.); (P.H.P.v.H.); (O.W.); (R.M.v.d.W.); (T.B.M.H.); (W.J.d.J.)
| | - Olaf Welting
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology and Metabolism, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, 1105 BK Amsterdam, The Netherlands; (S.R.); (P.H.P.v.H.); (O.W.); (R.M.v.d.W.); (T.B.M.H.); (W.J.d.J.)
| | - Hakim Rahaoui
- The Netherlands Organization for Applied Scientific Research (TNO), 3704 HE Zeist, The Netherlands; (H.R.); (F.S.)
| | - Frank Schuren
- The Netherlands Organization for Applied Scientific Research (TNO), 3704 HE Zeist, The Netherlands; (H.R.); (F.S.)
| | - Sybren L. Meijer
- Department of Pathology, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands;
| | - René M. van den Wijngaard
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology and Metabolism, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, 1105 BK Amsterdam, The Netherlands; (S.R.); (P.H.P.v.H.); (O.W.); (R.M.v.d.W.); (T.B.M.H.); (W.J.d.J.)
| | - Theodorus B. M. Hakvoort
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology and Metabolism, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, 1105 BK Amsterdam, The Netherlands; (S.R.); (P.H.P.v.H.); (O.W.); (R.M.v.d.W.); (T.B.M.H.); (W.J.d.J.)
| | - Wouter J. de Jonge
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology and Metabolism, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, 1105 BK Amsterdam, The Netherlands; (S.R.); (P.H.P.v.H.); (O.W.); (R.M.v.d.W.); (T.B.M.H.); (W.J.d.J.)
- Department of Surgery, University of Bonn, 53113 Bonn, Germany
| | - Sigrid E. M. Heinsbroek
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology and Metabolism, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, 1105 BK Amsterdam, The Netherlands; (S.R.); (P.H.P.v.H.); (O.W.); (R.M.v.d.W.); (T.B.M.H.); (W.J.d.J.)
| |
Collapse
|
52
|
Salami AT, Okotie GE, Echendu PN, Akpamu U, Olaleye SB. Potassium bromate (KBrO 3) modulates oxidative stress and inflammatory biomarkers in sodium hydroxide (NaOH) - induced Crohn's colitis in Wistar rats. Can J Physiol Pharmacol 2021; 99:989-999. [PMID: 33848442 DOI: 10.1139/cjpp-2020-0678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Potassium bromate (KBrO3) present in consumed ozonised water was recently documented to exacerbate experimental gastric ulcer. Information, however, is vague as regards its effects in the colon where water reabsorption occurs. In this study, we observed the possible effects of KBrO3 on oxidative stress and inflammatory biomarkers in sodium hydroxide (NaOH) - induced Crohn's colitis (CC). Wistar rats (180-200 g) were divided into six groups (n = 10): (i) control; (ii) untreated CC (induced by 1.4% NaOH; intra-rectal administration); and (iii-vi) CC treated with vitamin E, KBrO3, vitamin E+KBrO3, and sulphazalazine, respectively, for 7 days. Body weight and stool score were monitored daily. By day 3 and 7, excised colon was evaluated for ulcer scores and biochemical and histological analysis. Blood samples collected on days 3 and 7 were assayed for haematological indices using standard methods. Data were subjected to analysis of variance (ANOVA) and p ≤ 0.05 considered significant. Platelet/lymphocyte ratio, colonic ulcer score, malondialdehyde, and mast cells were significantly decreased while colonic sulfhydryl, and Ca2+- and Na+/K+-ATPase activities were increased following KBrO3 treatment compared with untreated CC. These findings suggest that KBrO3 may mitigate against NaOH-induced CC via inhibiting mast cell population and oxidative and inflammatory content but stimulating colonic sulfhydryl and Ca2+- and Na+/K+-ATPase activities.
Collapse
Affiliation(s)
- Adeola Temitope Salami
- Gastrointestinal secretions and inflammation research unit, Department of Physiology, University of Ibadan, Ibadan, Oyo State, Nigeria.,Gastrointestinal secretions and inflammation research unit, Department of Physiology, University of Ibadan, Ibadan, Oyo State, Nigeria
| | - Gloria Enevwo Okotie
- Gastrointestinal secretions and inflammation research unit, Department of Physiology, University of Ibadan, Ibadan, Oyo State, Nigeria.,Gastrointestinal secretions and inflammation research unit, Department of Physiology, University of Ibadan, Ibadan, Oyo State, Nigeria
| | - Precious Nekachi Echendu
- Gastrointestinal secretions and inflammation research unit, Department of Physiology, University of Ibadan, Ibadan, Oyo State, Nigeria.,Gastrointestinal secretions and inflammation research unit, Department of Physiology, University of Ibadan, Ibadan, Oyo State, Nigeria
| | - Uwaifoh Akpamu
- Gastrointestinal secretions and inflammation research unit, Department of Physiology, University of Ibadan, Ibadan, Oyo State, Nigeria.,Gastrointestinal secretions and inflammation research unit, Department of Physiology, University of Ibadan, Ibadan, Oyo State, Nigeria
| | - Samuel Babafemi Olaleye
- Gastrointestinal secretions and inflammation research unit, Department of Physiology, University of Ibadan, Ibadan, Oyo State, Nigeria.,Gastrointestinal secretions and inflammation research unit, Department of Physiology, University of Ibadan, Ibadan, Oyo State, Nigeria
| |
Collapse
|
53
|
Jang HJ, Son S, Kim JA, Jung MY, Choi YJ, Kim DH, Lee HK, Shin D, Kim Y. Characterization and Functional Test of Canine Probiotics. Front Microbiol 2021; 12:625562. [PMID: 33763044 PMCID: PMC7982664 DOI: 10.3389/fmicb.2021.625562] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 02/17/2021] [Indexed: 02/01/2023] Open
Abstract
Probiotics can modulate the composition of gut microbiota and benefit the host animal health in multiple ways. Lactic acid bacteria (LAB), mainly Lactobacillus and Bifidobacterium species, are well-known microbes with probiotic potential. In the present study, 88 microbial strains were isolated from canine feces and annotated. Among these, the four strains CACC517, 537, 558, and 566 were tested for probiotic characteristics, and their beneficial effects on hosts were evaluated both in vitro and in vivo; these strains exhibited antibiosis, antibiotic activity, acid and bile tolerance, and relative cell adhesion to the HT-29 monolayer cell line. Byproducts of these strains increased the viability and decreased oxidative stress in mouse and dog cell lines (RAW264.7 and DH82, respectively). Subsequently, when the probiotics were applied to the clinical trial, changes in microbial composition and relative abundance of bacterial strains were clearly observed in the experimental animals. Experimental groups before and after the application were obviously separated from PCA analysis of clinical results. Conclusively, these results could provide comprehensive understanding of the effects of probiotic strains (CACC517, 537, 558, and 566) and their industrial applications.
Collapse
Affiliation(s)
- Hyun-Jun Jang
- Department of Research and Development, Center for Industrialization of Agricultural and Livestock Microorganisms, Jeongeup-si, South Korea
| | - Seungwoo Son
- Department of Agricultural Convergence Technology, Jeonbuk National University, Jeonju-si, South Korea.,The Animal Molecular Genetics & Breeding Center, Jeonbuk National University, Jeonju-si, South Korea
| | - Jung-Ae Kim
- Department of Research and Development, Center for Industrialization of Agricultural and Livestock Microorganisms, Jeongeup-si, South Korea.,Department of Bioactive Material Sciences, Jeonbuk National University, Jeonju-si, South Korea
| | - Min Young Jung
- Department of Research and Development, Center for Industrialization of Agricultural and Livestock Microorganisms, Jeongeup-si, South Korea
| | - Yeon-Jae Choi
- Department of Research and Development, Center for Industrialization of Agricultural and Livestock Microorganisms, Jeongeup-si, South Korea
| | - Dae-Hyuk Kim
- Department of Research and Development, Center for Industrialization of Agricultural and Livestock Microorganisms, Jeongeup-si, South Korea.,Department of Bioactive Material Sciences, Jeonbuk National University, Jeonju-si, South Korea.,Department of Molecular Biology, Institute for Molecular Biology and Genetics, Jeonbuk National University, Jeonju-si, South Korea
| | - Hak Kyo Lee
- Department of Agricultural Convergence Technology, Jeonbuk National University, Jeonju-si, South Korea.,The Animal Molecular Genetics & Breeding Center, Jeonbuk National University, Jeonju-si, South Korea
| | - Donghyun Shin
- Department of Agricultural Convergence Technology, Jeonbuk National University, Jeonju-si, South Korea.,The Animal Molecular Genetics & Breeding Center, Jeonbuk National University, Jeonju-si, South Korea
| | - Yangseon Kim
- Department of Research and Development, Center for Industrialization of Agricultural and Livestock Microorganisms, Jeongeup-si, South Korea
| |
Collapse
|
54
|
Valter M, Verstockt S, Finalet Ferreiro JA, Cleynen I. Extracellular Vesicles in Inflammatory Bowel Disease: Small Particles, Big Players. J Crohns Colitis 2021; 15:499-510. [PMID: 32905585 DOI: 10.1093/ecco-jcc/jjaa179] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Extracellular vesicles are nanovesicles released by many cell types into the extracellular space. They are important mediators of intercellular communication, enabling the functional transfer of molecules from one cell to another. Moreover, their molecular composition reflects the physiological status of the producing cell and tissue. Consequently, these vesicles have been involved in many [patho]physiological processes such as immunomodulation and intestinal epithelial repair, both key processes involved in inflammatory bowel disease. Given that these vesicles are present in many body fluids, they also provide opportunities for diagnostic, prognostic, and therapeutic applications. In this review, we summarise functional roles of extracellular vesicles in health and disease, with a focus on immune regulation and intestinal barrier integrity, and review recent studies on extracellular vesicles and inflammatory bowel disease. We also elaborate on their clinical potential in inflammatory bowel disease.
Collapse
Affiliation(s)
- M Valter
- Laboratory for Complex Genetics, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - S Verstockt
- Translational Research center for Gastrointestinal Disorders [TARGID], Department of Chronic Diseases, Metabolism and Ageing [CHROMETA], KU Leuven, Leuven, Belgium
| | - J A Finalet Ferreiro
- Laboratory for Complex Genetics, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - I Cleynen
- Laboratory for Complex Genetics, Department of Human Genetics, KU Leuven, Leuven, Belgium
| |
Collapse
|
55
|
Engevik MA, Danhof HA, Ruan W, Engevik AC, Chang-Graham AL, Engevik KA, Shi Z, Zhao Y, Brand CK, Krystofiak ES, Venable S, Liu X, Hirschi KD, Hyser JM, Spinler JK, Britton RA, Versalovic J. Fusobacterium nucleatum Secretes Outer Membrane Vesicles and Promotes Intestinal Inflammation. mBio 2021; 12:e02706-20. [PMID: 33653893 PMCID: PMC8092269 DOI: 10.1128/mbio.02706-20] [Citation(s) in RCA: 108] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 01/22/2021] [Indexed: 12/17/2022] Open
Abstract
Multiple studies have implicated microbes in the development of inflammation, but the mechanisms remain unknown. Bacteria in the genus Fusobacterium have been identified in the intestinal mucosa of patients with digestive diseases; thus, we hypothesized that Fusobacterium nucleatum promotes intestinal inflammation. The addition of >50 kDa F. nucleatum conditioned media, which contain outer membrane vesicles (OMVs), to colonic epithelial cells stimulated secretion of the proinflammatory cytokines interleukin-8 (IL-8) and tumor necrosis factor (TNF). In addition, purified F. nucleatum OMVs, but not compounds <50 kDa, stimulated IL-8 and TNF production; which was decreased by pharmacological inhibition of Toll-like receptor 4 (TLR4). These effects were linked to downstream effectors p-ERK, p-CREB, and NF-κB. F. nucleatum >50-kDa compounds also stimulated TNF secretion, p-ERK, p-CREB, and NF-κB activation in human colonoid monolayers. In mice harboring a human microbiota, pretreatment with antibiotics and a single oral gavage of F. nucleatum resulted in inflammation. Compared to mice receiving vehicle control, mice treated with F. nucleatum showed disruption of the colonic architecture, with increased immune cell infiltration and depleted mucus layers. Analysis of mucosal gene expression revealed increased levels of proinflammatory cytokines (KC, TNF, IL-6, IFN-γ, and MCP-1) at day 3 and day 5 in F. nucleatum-treated mice compared to controls. These proinflammatory effects were absent in mice who received F. nucleatum without pretreatment with antibiotics, suggesting that an intact microbiome is protective against F. nucleatum-mediated immune responses. These data provide evidence that F. nucleatum promotes proinflammatory signaling cascades in the context of a depleted intestinal microbiome.IMPORTANCE Several studies have identified an increased abundance of Fusobacterium in the intestinal tracts of patients with colon cancer, liver cirrhosis, primary sclerosing cholangitis, gastroesophageal reflux disease, HIV infection, and alcoholism. However, the direct mechanism(s) of action of Fusobacterium on pathophysiological within the gastrointestinal tract is unclear. These studies have identified that F. nucleatum subsp. polymorphum releases outer membrane vesicles which activate TLR4 and NF-κB to stimulate proinflammatory signals in vitro Using mice harboring a human microbiome, we demonstrate that F. nucleatum can promote inflammation, an effect which required antibiotic-mediated alterations in the gut microbiome. Collectively, these results suggest a mechanism by which F. nucleatum may contribute to intestinal inflammation.
Collapse
Affiliation(s)
- Melinda A Engevik
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, USA
- Department of Pathology, Texas Children's Hospital, Houston, Texas, USA
| | - Heather A Danhof
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Wenly Ruan
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Amy C Engevik
- Department of Surgical Sciences, Vanderbilt University Medical Center, Nashville Tennessee, USA
| | - Alexandra L Chang-Graham
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Kristen A Engevik
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Zhongcheng Shi
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, USA
- Department of Pathology, Texas Children's Hospital, Houston, Texas, USA
| | - Yanling Zhao
- Department of Pediatrics, Texas Children's Cancer Center, Texas Children's Hospital, Houston, Texas, USA
| | - Colleen K Brand
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Evan S Krystofiak
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, USA
| | - Susan Venable
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, USA
- Department of Pathology, Texas Children's Hospital, Houston, Texas, USA
| | - Xinli Liu
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, Texas, USA
| | - Kendal D Hirschi
- Department of Pediatrics and Human and Molecular Genetics, Children's Nutrition Research Center, Baylor College of Medicine, Houston, Texas, USA
| | - Joseph M Hyser
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Jennifer K Spinler
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, USA
- Department of Pathology, Texas Children's Hospital, Houston, Texas, USA
| | - Robert A Britton
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - James Versalovic
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, USA
- Department of Pathology, Texas Children's Hospital, Houston, Texas, USA
| |
Collapse
|
56
|
Deng L, Wojciech L, Gascoigne NRJ, Peng G, Tan KSW. New insights into the interactions between Blastocystis, the gut microbiota, and host immunity. PLoS Pathog 2021; 17:e1009253. [PMID: 33630979 PMCID: PMC7906322 DOI: 10.1371/journal.ppat.1009253] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The human gut microbiota is a diverse and complex ecosystem that is involved in beneficial physiological functions as well as disease pathogenesis. Blastocystis is a common protistan parasite and is increasingly recognized as an important component of the gut microbiota. The correlations between Blastocystis and other communities of intestinal microbiota have been investigated, and, to a lesser extent, the role of this parasite in maintaining the host immunological homeostasis. Despite recent studies suggesting that Blastocystis decreases the abundance of beneficial bacteria, most reports indicate that Blastocystis is a common component of the healthy gut microbiome. This review covers recent finding on the potential interactions between Blastocystis and the gut microbiota communities and its roles in regulating host immune responses.
Collapse
Affiliation(s)
- Lei Deng
- Laboratory of Molecular and Cellular Parasitology, Healthy Aging Programme and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Lukasz Wojciech
- Immunology Programme and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Nicholas R. J. Gascoigne
- Immunology Programme and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Guangneng Peng
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Kevin S. W. Tan
- Laboratory of Molecular and Cellular Parasitology, Healthy Aging Programme and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
57
|
Agrawal G, Aitken J, Hamblin H, Collins M, Borody TJ. Putting Crohn's on the MAP: Five Common Questions on the Contribution of Mycobacterium avium subspecies paratuberculosis to the Pathophysiology of Crohn's Disease. Dig Dis Sci 2021; 66:348-358. [PMID: 33089484 PMCID: PMC7577843 DOI: 10.1007/s10620-020-06653-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/02/2020] [Indexed: 02/06/2023]
Abstract
For decades, Mycobacterium avium subspecies paratuberculosis (MAP) has been linked to the pathogenesis of Crohn's disease. Despite many investigations and research efforts, there remains no clear unifying explanation of its pathogenicity to humans. Proponents argue Crohn's disease shares many identical features with a granulomatous infection in ruminants termed Johne's disease and similarities with ileo-cecal tuberculosis. Both are caused by species within the Mycobacterium genus. Sceptics assert that since MAP is found in individuals diagnosed with Crohn's disease as well as in healthy population controls, any association with CD is coincidental. This view is supported by the uncertain response of patients to antimicrobial therapy. This report aims to address the controversial aspects of this proposition with information and knowledge gathered from several disciplines, including microbiology and veterinary medicine. The authors hope that this discussion will stimulate further research aimed at confirming or refuting the contribution of MAP to the pathogenesis of Crohn's disease and ultimately lead to advanced targeted clinical therapies.
Collapse
Affiliation(s)
- Gaurav Agrawal
- Gastroenterology and Infectious Diseases, Centre for Digestive Diseases, Sydney, Australia.
- Guy's and St. Thomas' NHS Foundation Trust & King's College, London, UK.
| | - John Aitken
- Microbiology, Otakaro Pathways, Christchurch, New Zealand
| | - Harrison Hamblin
- Gastroenterology and Infectious Diseases, Centre for Digestive Diseases, Sydney, Australia
| | - Michael Collins
- Veterinary Microbiology, Department of Pathobiological Sciences, University of Wisconsin, Madison, USA
| | - Thomas J Borody
- Gastroenterology and Infectious Diseases, Centre for Digestive Diseases, Sydney, Australia
| |
Collapse
|
58
|
de Castro MM, Pascoal LB, Steigleder KM, Siqueira BP, Corona LP, Ayrizono MDLS, Milanski M, Leal RF. Role of diet and nutrition in inflammatory bowel disease. World J Exp Med 2021; 11:1-16. [PMID: 33585174 PMCID: PMC7852575 DOI: 10.5493/wjem.v11.i1.1] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 11/02/2020] [Accepted: 11/12/2020] [Indexed: 02/06/2023] Open
Abstract
Inflammatory bowel diseases (IBDs) are closely linked to nutrition. The latest research indicates that diet and nutrition are significantly involved in the etiopathogenesis of the disease, although their specific role throughout its clinical course still remains unclear. This study reviewed how diet and nutrition are associated with IBD development and management. Even though specific diets have been shown to bring about positive outcomes, there is currently no scientific consensus regarding an appropriate diet that would benefit all IBD patients. We suggest that individualized dietary recommendations are of the greatest importance and that diets should be planned to provide individual IBD patients with specific nutrient requirements while keeping all the clinical aspects of the patients in mind. Further research is clearly necessary to investigate nutritional factors involved in IBD development and, especially, to evaluate the applications of the diets during the course of the disease.
Collapse
Affiliation(s)
- Marina Moreira de Castro
- IBD Research Laboratory, Colorectal Surgery Unit, School of Medical Sciences, University of Campinas (UNICAMP), Campinas 13083-878, São Paulo, Brazil
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas (UNICAMP), Campinas 13083-878, São Paulo, Brazil
| | - Lívia Bitencourt Pascoal
- IBD Research Laboratory, Colorectal Surgery Unit, School of Medical Sciences, University of Campinas (UNICAMP), Campinas 13083-878, São Paulo, Brazil
| | - Karine Mariane Steigleder
- IBD Research Laboratory, Colorectal Surgery Unit, School of Medical Sciences, University of Campinas (UNICAMP), Campinas 13083-878, São Paulo, Brazil
| | - Beatriz Piatezzi Siqueira
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas (UNICAMP), Limeira 13484-350, São Paulo, Brazil
| | - Ligiana Pires Corona
- Laboratory of Nutritional Epidemiology, School of Applied Sciences, University of Campinas (UNICAMP), Limeira 13484-350, São Paulo, Brazil
| | - Maria de Lourdes Setsuko Ayrizono
- IBD Research Laboratory, Colorectal Surgery Unit, School of Medical Sciences, University of Campinas (UNICAMP), Campinas 13083-878, São Paulo, Brazil
| | - Marciane Milanski
- IBD Research Laboratory, Colorectal Surgery Unit, School of Medical Sciences, University of Campinas (UNICAMP), Campinas 13083-878, São Paulo, Brazil
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas (UNICAMP), Campinas 13083-878, São Paulo, Brazil
| | - Raquel Franco Leal
- IBD Research Laboratory, Colorectal Surgery Unit, School of Medical Sciences, University of Campinas (UNICAMP), Campinas 13083-878, São Paulo, Brazil
| |
Collapse
|
59
|
Castleman MJ, Dillon SM, Purba C, Cogswell AC, McCarter M, Barker E, Wilson C. Enteric bacteria induce IFNγ and Granzyme B from human colonic Group 1 Innate Lymphoid Cells. Gut Microbes 2020; 12:1667723. [PMID: 31583949 PMCID: PMC7524156 DOI: 10.1080/19490976.2019.1667723] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Group 1 Innate Lymphoid Cells (which include Natural Killer cells and ILC1s) aid in gut anti-bacterial defense through the production of IFNγ, which is critical for mobilizing protective responses against enteric pathogens. When intestinal epithelial barrier integrity is compromised, commensal bacteria are likely to translocate from the gut lumen into the lamina propria. Few studies have addressed the mechanisms by which commensal bacteria impact the function of gut Group 1 ILCs, especially ILC1s. Utilizing an in vitro human colonic lamina propria mononuclear cell (LPMC) model, we evaluated Group 1 ILC cytokine and cytolytic protein production in response to a panel of enteric Gram-positive and Gram-negative commensal and pathogenic bacteria. IFNγ-production by NK cells and ILC1s was significantly increased after LPMC exposure to Gram-negative commensal or pathogenic bacteria, but not after exposure to the Gram-positive bacteria commensals tested. Stimulation of IFNγ production from Group 1 ILCs was not through direct recognition of bacteria by NK cells or ILC1s, but rather required accessory cells within the LPMC population. Myeloid dendritic cells generated IL-12p70, IL-18, and IL-1β upon exposure to enteric bacteria and these cytokines contributed to Group 1 ILC production of IFNγ. Furthermore, Gram-negative commensal or pathogenic bacteria induced significant expression of Granzyme B in NK cells and ILC1s. Overall, these data demonstrate that some enteric commensal bacteria indirectly induce inflammatory cytokine production and cytolytic protein expression from human colonic Group 1 ILCs, a process which could contribute to inflammation in the setting of microbial translocation.
Collapse
Affiliation(s)
- Moriah J. Castleman
- Department of Medicine, Division of Infectious Disease, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Stephanie M. Dillon
- Department of Medicine, Division of Infectious Disease, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Christine Purba
- Department of Medicine, Division of Infectious Disease, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Andrew C. Cogswell
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL, USA
| | - Martin McCarter
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Edward Barker
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL, USA
| | - Cara Wilson
- Department of Medicine, Division of Infectious Disease, University of Colorado Anschutz Medical Campus, Aurora, CO, USA,CONTACT Cara Wilson Department of Medicine, Division of Infectious Disease, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
60
|
Guilin Z, Pengyu Z, Wei L, Fengqi H, Chen F, Yu Y, Yue H, Yuting T. Reduction of gut microbial diversity and short chain fatty acids in BALB/c mice exposure to microcystin-LR. ECOTOXICOLOGY (LONDON, ENGLAND) 2020; 29:1347-1357. [PMID: 32772242 DOI: 10.1007/s10646-020-02254-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 07/09/2020] [Indexed: 06/11/2023]
Abstract
Gut microbiota has been shown to play critical roles in host health. The present study was to determine the toxicological effects of microcystin-LR (MCLR) on gut microbial community and metabolites using 16S rDNA sequencing and gas chromatography-mass spectrometry (GC-MS). MCLR was administered to BALB/c mice by gavage for eight weeks. Results of the microbial alpha-diversity (Sobs, Chao1, ACE and Shannon indexes) decreased in MCLR-treated group versus controls. Phylum Candidatus Saccharibacteria decreased significantly in MCLR-treated group versus controls. Correspondingly, more than thirties genera in relative abundance decreased, especially short chain fatty acid (SCFA)-producing bacteria (e.g., Alistipes and Ruminococcus). These results indicated that the gut microbial community structure was remarkably changed by MCLR. Furthermore, concentrations of SCFAs were significantly decreased after MCLR exposure (P < 0.01), where butyrate decreased as high as 4.9-fold. Consequently, sub-chronic exposure to MCLR could not only alter the microbial composition but metabolites. This study offered novel insights into the toxic mechanism of MCs from gut microbiota, and facilitated further clarification of risks to human health from MCs exposure.
Collapse
Affiliation(s)
- Zhang Guilin
- Changchun University of Science and Technology, School of Life Science and Technology, Changchun, 130022, China
| | - Zhang Pengyu
- Changchun University of Science and Technology, School of Life Science and Technology, Changchun, 130022, China
| | - Li Wei
- Changchun University of Science and Technology, School of Life Science and Technology, Changchun, 130022, China.
- Wuhan Haidian Foreign Language Shi Yan School, Wuhan, 430220, China.
| | - Hao Fengqi
- Changchun University of Science and Technology, School of Life Science and Technology, Changchun, 130022, China
| | - Fangmei Chen
- Changchun University of Science and Technology, School of Life Science and Technology, Changchun, 130022, China
| | - Yang Yu
- Changchun University of Science and Technology, School of Life Science and Technology, Changchun, 130022, China
| | - Hou Yue
- Changchun University of Science and Technology, School of Life Science and Technology, Changchun, 130022, China
| | - Tao Yuting
- Changchun University of Science and Technology, School of Life Science and Technology, Changchun, 130022, China
| |
Collapse
|
61
|
Castañeda S, Muñoz M, Villamizar X, Hernández PC, Vásquez LR, Tito RY, Ramírez JD. Microbiota characterization in Blastocystis-colonized and Blastocystis-free school-age children from Colombia. Parasit Vectors 2020; 13:521. [PMID: 33066814 PMCID: PMC7565366 DOI: 10.1186/s13071-020-04392-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 10/07/2020] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Blastocystis is a protist that lives in the intestinal tract of a variety of hosts, including humans. It is still unclear how Blastocystis causes disease, which presents an ongoing challenge for researchers. Despite the controversial findings on the association between Blastocystis and clinical digestive manifestations, there is currently no consensus as to whether this protozoan actually behaves as a pathogen in humans. Furthermore, the relationship between Blastocystis and the intestinal microbiota composition is not yet clear. For that reason, the aim of this study was to identify if colonization by Blastocystis is related to changes in the diversity and relative abundance of bacterial communities, compared with those of Blastocystis-free individuals in a group of Colombian children. METHODS We took stool samples from 57 school-aged children attending a daycare institution in Popayán (Southwest Colombia). Whole DNA was extracted and examined by 16S-rRNA amplicon-based sequencing. Blastocystis was detected by real time PCR and other intestinal parasites were detected by microscopy. We evaluated if Blastocystis was associated with host variables and the diversity and abundance of microbial communities. RESULTS The composition of the intestinal bacterial community was not significantly different between Blastocystis-free and Blastocystis-colonized children. Despite this, we observed a higher microbial richness in the intestines of children colonized by Blastocystis, which could, therefore, be considered a benefit to intestinal health. The phylum Firmicutes was the predominant taxonomic unit in both groups analyzed. In Blastocystis-free individuals, there was a higher proportion of Bacteroidetes; similarly, in children colonized by Blastocystis, there was a higher relative abundance of the phylum Proteobacteria; however, no statistically significant differences were found between the comparison groups. CONCLUSIONS The presence of Blastocystis showed a decrease in Bacteroides, and an increase in the relative abundance of the genus Faecalibacterium. It was also evident that the presence of Blastocystis was unrelated to dysbiosis at the intestinal level; on the contrary, its presence did not show statistically differences in the intestinal microbiota composition. Nevertheless, we believe that Blastocystis plays a role in the ecology of the intestinal microbiota through its interaction with other microbial components.
Collapse
Affiliation(s)
- Sergio Castañeda
- Grupo de Investigaciones Microbiológicas-UR (GIMUR), Departamento de Biología, Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
- Maestría en Informática Biomédica, Facultad de Medicina, Universidad El Bosque, Bogotá, Colombia
| | - Marina Muñoz
- Grupo de Investigaciones Microbiológicas-UR (GIMUR), Departamento de Biología, Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
| | - Ximena Villamizar
- Grupo de Investigaciones Microbiológicas-UR (GIMUR), Departamento de Biología, Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
| | - Paula C Hernández
- Laboratorio de Parasitología Molecular, Vicerrectoría de Investigaciones, Universidad El Bosque, Bogotá, Colombia
| | - Luis Reinel Vásquez
- Centro de Estudios en Microbiología y Parasitología (CEMPA), Departamento de Medicina Interna, Facultad de Ciencias de la Salud, Universidad del Cauca, Popayán, Colombia
| | - Raúl Yhossef Tito
- Department of Microbiology and Immunology, Rega Institute, KU Leuven, University of Leuven, Leuven, Belgium
| | - Juan David Ramírez
- Grupo de Investigaciones Microbiológicas-UR (GIMUR), Departamento de Biología, Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia.
| |
Collapse
|
62
|
Cai TT, Ye XL, Li RR, Chen H, Wang YY, Yong HJ, Pan ML, Lu W, Tang Y, Miao H, Snijders AM, Mao JH, Liu XY, Lu YB, Ding DF. Resveratrol Modulates the Gut Microbiota and Inflammation to Protect Against Diabetic Nephropathy in Mice. Front Pharmacol 2020; 11:1249. [PMID: 32973502 PMCID: PMC7466761 DOI: 10.3389/fphar.2020.01249] [Citation(s) in RCA: 109] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 07/29/2020] [Indexed: 12/18/2022] Open
Abstract
Oral administration of resveratrol is able to ameliorate the progression of diabetic nephropathy (DN); however, its mechanisms of action remain unclear. Recent evidence suggested that the gut microbiota is involved in the metabolism therapeutics. In the current study, we sought to determine whether the anti-DN effects of resveratrol are mediated through modulation of the gut microbiota using the genetic db/db mouse model of DN. We demonstrate that resveratrol treatment of db/db mice relieves a series of clinical indicators of DN. We then show that resveratrol improves intestinal barrier function and ameliorates intestinal permeability and inflammation. The composition of the gut microbiome was significantly altered in db/db mice compared to control db/m mice. Dysbiosis in db/db mice characterized by low abundance levels of Bacteroides, Alistipes, Rikenella, Odoribacter, Parabacteroides, and Alloprevotella genera were reversed by resveratrol treatment, suggesting a potential role for the microbiome in DN progression. Furthermore, fecal microbiota transplantation, derived from healthy resveratrol-treated db/m mice, was sufficient to antagonize the renal dysfunction, rebalance the gut microbiome and improve intestinal permeability and inflammation in recipient db/db mice. These results indicate that resveratrol-mediated changes in the gut microbiome may play an important role in the mechanism of action of resveratrol, which provides supporting evidence for the gut–kidney axis in DN.
Collapse
Affiliation(s)
- Ting-Ting Cai
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiao-Long Ye
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ru-Run Li
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hui Chen
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ya-Yun Wang
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hui-Juan Yong
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ming-Lin Pan
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wei Lu
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ying Tang
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Heng Miao
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Antoine M Snijders
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, United States
| | - Jian-Hua Mao
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, United States
| | - Xing-Yin Liu
- Department of Pathogen Biology-Microbiology Division, Key Laboratory of Pathogen of Jiangsu Province, Nanjing Medical University, Nanjing, China
| | - Yi-Bing Lu
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Da-Fa Ding
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
63
|
Abstract
The last decade has witnessed unparalleled advances in our understanding of the complexity of the oral microbiome and the compositional changes that occur in subgingival biofilms in the transition from health to gingivitis and to destructive periodontal disease. The traditional view, which has held sway for the last 2 decades, that disease is characterized by the outgrowth of a consortium, or consortia, of a limited number of potentially pathogenic organisms, has given way to an alternative paradigm. In this new view, the microbiological changes associated with disease represent whole-scale alterations to the overall microbial population structure and to the functional properties of the entire community. Thus, and in common with other microbially mediated diseases of the gastrointestinal tract, the normally balanced, symbiotic, and generally benign commensal microbiome of the tooth-associated biofilm undergoes dysbiosis to a potentially deleterious microbiota. Coincident with progress in defining the microbiology of these diseases, there have been equally important advances in our understanding of the inflammatory systems of the periodontal tissues, their control, and how inflammation may contribute both to the development of dysbiosis and, in a deregulated state, the destructive disease process. One can therefore speculate that the inflammatory response and the periodontal microbiome are in a bidirectional balance in oral health and a bidirectional imbalance in periodontitis. However, despite these clear insights into both sides of the host/microbe balance in periodontal disease, there remain several unresolved issues concerning the role of the microbiota in disease. These include, but are not limited to, the factors which determine progression from gingivitis to periodontitis in a proportion of the population, whether dysbiosis causes disease or results from disease, and the molecular details of the microbial stimulus responsible for driving the destructive inflammatory response. Further progress in resolving these issues may provide significant benefit to diagnosis, treatment, and prevention.
Collapse
Affiliation(s)
- Mike A Curtis
- Centre for Host Microbiome Interactions, Faculty of Dentistry, Oral& Craniofacial Sciences, King's College London, London, UK
| | - Patricia I Diaz
- School of Dental Medicine, UConn Health, Farmington, Connecticut, USA
| | - Thomas E Van Dyke
- Department of Applied Oral Sciences, The Forsyth Institute, Cambridge, Massachusetts, USA
| |
Collapse
|
64
|
Cui Z, Wu S, Li J, Yang QE, Chai S, Wang L, Wang X, Zhang X, Liu S, Yao J. Effect of Alfalfa Hay and Starter Feeding Intervention on Gastrointestinal Microbial Community, Growth and Immune Performance of Yak Calves. Front Microbiol 2020; 11:994. [PMID: 32582049 PMCID: PMC7287295 DOI: 10.3389/fmicb.2020.00994] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 04/23/2020] [Indexed: 12/21/2022] Open
Abstract
The present study aims to evaluate the effects of different early weaning paradigms, which supplied with extra alfalfa hay, or starter feeding, or both alfalfa hay and starter feeding, along with the milk replacer, on the gastrointestinal microbial community, growth, and immune performance of yak calves. Twenty 30-day-old male yak calves were randomly assigned to four groups, including the control (CON), alfalfa hay (A), starter feeding (S), and starter plus alfalfa hay (SA) groups. The gastrointestinal microbial colonization, the gastrointestinal development and function, and the growth and immune performance of all the yak calves were separately measured. Supplementation with alfalfa and starter feeding during the pre-weaning period significantly increased body weight, body height, body length, and chest girth. The significantly improved rumen fermentation and promoted intestinal digestion-absorption function in alfalfa and starter feeding groups, including the identified significantly increased concentrations of ruminal total volatile fatty acid (VFA); the significantly increased concentrations and proportions of acetate, butyrate, and isovalerate; the increased α-amylase activities in the duodenum, jejunum, and ileum; the increased papillae length and width of rumen epithelium and rumen wall thickness; and the increased villus height and crypt depth of the duodenum, jejunum, and ileum, could all contribute to promote the growth of calves. These significant improvements on rumen fermentation and intestinal digestion-absorption function could be further attributed to the increased proliferation of starch-decomposing, and cellulose- or hemicellulose-decomposing bacteria identified in the rumen, jejunum, and ileum. Furthermore, based on the expression of intestinal inflammatory cytokines and the rumen epithelial RNA sequencing results, alfalfa supplementation reduced the occurrence of ruminal and intestinal inflammation, whereas starter feeding supplementation was mainly beneficial to the differentiation of immune cells and the improved immune function. Meanwhile, the significantly altered relative abundances of genera in the SA group, including increased relative abundance of Limnobacter, Escherichia/Shigella, and Aquabacterium in the rumen and increased relative abundance of Coprococcus, Pseudobutyrivibrio, Flavonifractor, Synergistes, and Sutterella in jejunum, were able to reduce gastrointestinal inflammation and enhance the immune function, which enhanced the immune function of the yak calves fed with alfalfa and starter feeding. Overall, milk replacer supplemented with alfalfa and starter feeding during the pre-weaning period could alter gastrointestinal microbiota and then benefit the gastrointestinal development, digestion-absorption function, growth, and immune performance of the yak calves.
Collapse
Affiliation(s)
- Zhanhong Cui
- Qinghai Academy of Animal Science and Veterinary Medicine, Qinghai University, Xining, China.,College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Shengru Wu
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Jilan Li
- Qinghai Academy of Animal Science and Veterinary Medicine, Qinghai University, Xining, China
| | - Qi-En Yang
- Northwest Plateau Institute of Biology, Chinese Academy of Sciences, Xining, China
| | - Shatuo Chai
- Qinghai Academy of Animal Science and Veterinary Medicine, Qinghai University, Xining, China
| | - Lei Wang
- Qinghai Academy of Animal Science and Veterinary Medicine, Qinghai University, Xining, China
| | - Xun Wang
- Qinghai Academy of Animal Science and Veterinary Medicine, Qinghai University, Xining, China
| | - Xiaowei Zhang
- Qinghai Academy of Animal Science and Veterinary Medicine, Qinghai University, Xining, China
| | - Shujie Liu
- Qinghai Academy of Animal Science and Veterinary Medicine, Qinghai University, Xining, China
| | - Junhu Yao
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| |
Collapse
|
65
|
Andrighetti T, Bohar B, Lemke N, Sudhakar P, Korcsmaros T. MicrobioLink: An Integrated Computational Pipeline to Infer Functional Effects of Microbiome-Host Interactions. Cells 2020; 9:cells9051278. [PMID: 32455748 PMCID: PMC7291277 DOI: 10.3390/cells9051278] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/15/2020] [Accepted: 05/19/2020] [Indexed: 02/07/2023] Open
Abstract
Microbiome–host interactions play significant roles in health and in various diseases including autoimmune disorders. Uncovering these inter-kingdom cross-talks propels our understanding of disease pathogenesis and provides useful leads on potential therapeutic targets. Despite the biological significance of microbe–host interactions, there is a big gap in understanding the downstream effects of these interactions on host processes. Computational methods are expected to fill this gap by generating, integrating, and prioritizing predictions—as experimental detection remains challenging due to feasibility issues. Here, we present MicrobioLink, a computational pipeline to integrate predicted interactions between microbial and host proteins together with host molecular networks. Using the concept of network diffusion, MicrobioLink can analyse how microbial proteins in a certain context are influencing cellular processes by modulating gene or protein expression. We demonstrated the applicability of the pipeline using a case study. We used gut metaproteomic data from Crohn’s disease patients and healthy controls to uncover the mechanisms by which the microbial proteins can modulate host genes which belong to biological processes implicated in disease pathogenesis. MicrobioLink, which is agnostic of the microbial protein sources (bacterial, viral, etc.), is freely available on GitHub.
Collapse
Affiliation(s)
- Tahila Andrighetti
- Earlham Institute, Norwich Research Park, Norwich NR4 7UZ, UK; (T.A.); (B.B.)
- Institute of Biosciences, São Paulo University (UNESP), Botucatu 18618-689, SP, Brazil;
| | - Balazs Bohar
- Earlham Institute, Norwich Research Park, Norwich NR4 7UZ, UK; (T.A.); (B.B.)
- Department of Genetics, Eötvös Loránd University, Budapest 1117, Hungary
| | - Ney Lemke
- Institute of Biosciences, São Paulo University (UNESP), Botucatu 18618-689, SP, Brazil;
| | - Padhmanand Sudhakar
- Earlham Institute, Norwich Research Park, Norwich NR4 7UZ, UK; (T.A.); (B.B.)
- Quadram Institute Bioscience, Norwich Research Park, Norwich NR4 7UQ, UK
- Department of Chronic Diseases, Metabolism and Ageing, KU Leuven BE-3000, Leuven, Belgium
- Correspondence: (T.K.); (P.S.)
| | - Tamas Korcsmaros
- Earlham Institute, Norwich Research Park, Norwich NR4 7UZ, UK; (T.A.); (B.B.)
- Quadram Institute Bioscience, Norwich Research Park, Norwich NR4 7UQ, UK
- Correspondence: (T.K.); (P.S.)
| |
Collapse
|
66
|
Schanz O, Chijiiwa R, Cengiz SC, Majlesain Y, Weighardt H, Takeyama H, Förster I. Dietary AhR Ligands Regulate AhRR Expression in Intestinal Immune Cells and Intestinal Microbiota Composition. Int J Mol Sci 2020; 21:ijms21093189. [PMID: 32366032 PMCID: PMC7246938 DOI: 10.3390/ijms21093189] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/24/2020] [Accepted: 04/28/2020] [Indexed: 12/12/2022] Open
Abstract
A diet rich in vegetables and fruit is generally considered healthy because of a high content of phytochemicals, vitamins, and fiber. The phytochemical indole-3-carbinol (I3C), a derivative of glucobrassicin, is sold as a dietary supplement promising diverse health benefits. I3C metabolites act as ligands of the aryl hydrocarbon receptor (AhR), an important sensor for environmental polyaromatic chemicals. Here, we investigated how dietary AhR ligand supplementation influences AhR target gene expression and intestinal microbiota composition. For this, we used AhR repressor (AhRR)-reporter mice as a tool to study AhR activation in the intestine following dietary I3C-supplementation in comparison with AhR ligand-deprived diets, including a high fat diet. AhRR expression in intestinal immune cells was mainly driven by dietary AhR ligands and was independent of microbial metabolites. A lack of dietary AhR ligands caused enhanced susceptibility to dextran sodium sulfate (DSS)-induced colitis and correlated with the expansion of Enterobacteriaceae, whereas Clostridiales, Muribaculaceae, and Rikenellaceae were strongly reduced. I3C supplementation largely reverted this effect. Comparison of I3C-induced changes in microbiota composition using wild-type (WT), AhRR-deficient, and AhR-deficient mice revealed both AhR-dependent and -independent alterations in the microbiome. Overall, our study demonstrates that dietary AhR ligand supplementation has a profound influence on Ahrr expression in intestinal immune cells as well as microbiota composition.
Collapse
Affiliation(s)
- Oliver Schanz
- Immunology and Environment, Life and Medical Sciences (LIMES) Institute, University of Bonn, Carl-Troll-Straße 31, 53115 Bonn, Germany; (O.S.); (S.C.C.); (Y.M.)
| | - Rieka Chijiiwa
- Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University, 2-2 Wakamatsucho, Shinjuku-ku, Tokyo 162-8480, Japan;
- Computational Bio Big-Data Open Innovation Laboratory (CBBD-OIL), National Institute of Advanced Industrial Science and Technology, 3-4-1 Okubo, Shinjuku-ku, Tokyo 169-8555, Japan
| | - Sevgi Can Cengiz
- Immunology and Environment, Life and Medical Sciences (LIMES) Institute, University of Bonn, Carl-Troll-Straße 31, 53115 Bonn, Germany; (O.S.); (S.C.C.); (Y.M.)
| | - Yasmin Majlesain
- Immunology and Environment, Life and Medical Sciences (LIMES) Institute, University of Bonn, Carl-Troll-Straße 31, 53115 Bonn, Germany; (O.S.); (S.C.C.); (Y.M.)
| | - Heike Weighardt
- Immunology and Environment, Life and Medical Sciences (LIMES) Institute, University of Bonn, Carl-Troll-Straße 31, 53115 Bonn, Germany; (O.S.); (S.C.C.); (Y.M.)
- Correspondence: (H.W.); (H.T.); (I.F.); Tel.: +49-228-73-62706 (H.W.); +81-3-5369-7326 (H.T.); +49-228-73-62780 (I.F.)
| | - Haruko Takeyama
- Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University, 2-2 Wakamatsucho, Shinjuku-ku, Tokyo 162-8480, Japan;
- Computational Bio Big-Data Open Innovation Laboratory (CBBD-OIL), National Institute of Advanced Industrial Science and Technology, 3-4-1 Okubo, Shinjuku-ku, Tokyo 169-8555, Japan
- Research Organization for Nano and Life Innovation, Waseda University, 513 Wasedatsurumaki-cho, Shinjuku-ku, Tokyo 162-0041, Japan
- Institute for Advanced Research of Biosystem Dynamics, Waseda Research Institute for Science and Engineering, Graduate School of Advanced Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo 169-8555, Japan
- Correspondence: (H.W.); (H.T.); (I.F.); Tel.: +49-228-73-62706 (H.W.); +81-3-5369-7326 (H.T.); +49-228-73-62780 (I.F.)
| | - Irmgard Förster
- Immunology and Environment, Life and Medical Sciences (LIMES) Institute, University of Bonn, Carl-Troll-Straße 31, 53115 Bonn, Germany; (O.S.); (S.C.C.); (Y.M.)
- Correspondence: (H.W.); (H.T.); (I.F.); Tel.: +49-228-73-62706 (H.W.); +81-3-5369-7326 (H.T.); +49-228-73-62780 (I.F.)
| |
Collapse
|
67
|
Schepper JD, Collins F, Rios-Arce ND, Kang HJ, Schaefer L, Gardinier JD, Raghuvanshi R, Quinn RA, Britton R, Parameswaran N, McCabe LR. Involvement of the Gut Microbiota and Barrier Function in Glucocorticoid-Induced Osteoporosis. J Bone Miner Res 2020; 35:801-820. [PMID: 31886921 DOI: 10.1002/jbmr.3947] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 12/05/2019] [Accepted: 12/14/2019] [Indexed: 12/14/2022]
Abstract
Glucocorticoids (GCs) are potent immune-modulating drugs with significant side effects, including glucocorticoid-induced osteoporosis (GIO). GCs directly induce osteoblast and osteocyte apoptosis but also alter intestinal microbiota composition. Although the gut microbiota is known to contribute to the regulation of bone density, its role in GIO has never been examined. To test this, male C57/Bl6J mice were treated for 8 weeks with GC (prednisolone, GC-Tx) in the presence or absence of broad-spectrum antibiotic treatment (ABX) to deplete the microbiota. Long-term ABX prevented GC-Tx-induced trabecular bone loss, showing the requirement of gut microbiota for GIO. Treatment of GC-Tx mice with a probiotic (Lactobacillus reuteri [LR]) prevented trabecular bone loss. Microbiota analyses indicated that GC-Tx changed the abundance of Verrucomicobiales and Bacteriodales phyla and random forest analyses indicated significant differences in abundance of Porphyromonadaceae and Clostridiales operational taxonomic units (OTUs) between groups. Furthermore, transplantation of GC-Tx mouse fecal material into recipient naïve, untreated WT mice caused bone loss, supporting a functional role for microbiota in GIO. We also report that GC caused intestinal barrier breaks, as evidenced by increased serum endotoxin level (2.4-fold), that were prevented by LR and ABX treatments. Enhancement of barrier function with a mucus supplement prevented both GC-Tx-induced barrier leakage and trabecular GIO. In bone, treatment with ABX, LR or a mucus supplement reduced GC-Tx-induced osteoblast and osteocyte apoptosis. GC-Tx suppression of Wnt10b in bone was restored by the LR and high-molecular-weight polymer (MDY) treatments as well as microbiota depletion. Finally, we identified that bone-specific Wnt10b overexpression prevented GIO. Taken together, our data highlight the previously unappreciated involvement of the gut microbiota and intestinal barrier function in trabecular GIO pathogenesis (including Wnt10b suppression and osteoblast and osteocyte apoptosis) and identify the gut as a novel therapeutic target for preventing GIO. © 2019 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
| | - Fraser Collins
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Naiomy Deliz Rios-Arce
- Department of Physiology, Michigan State University, East Lansing, MI, USA.,Comparative Medicine and Integrative Biology Program, Michigan State University, East Lansing, MI, USA
| | - Ho Jun Kang
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Laura Schaefer
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | | | - Ruma Raghuvanshi
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA
| | - Robert A Quinn
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA
| | - Robert Britton
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | | | - Laura R McCabe
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
68
|
Sugiyama T, Sasaki M, Nakagawa S, Inoue S, Adachi K, Yoshimine T, Yamaguchi Y, Tamura Y, Izawa S, Hijikata Y, Ebi M, Mizuno M, Yamamoto S, Funaki Y, Ogasawara N, Goto C, Kasugai K. The association among enterobacterial flora, dietary factors, and prognosis in patients with ulcerative colitis. J Clin Biochem Nutr 2020; 66:152-157. [PMID: 32231412 DOI: 10.3164/jcbn.19-90] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 12/04/2019] [Indexed: 12/16/2022] Open
Abstract
The role of enterobacterial flora in the onset and progression of inflammatory bowel diseases is a topic of considerable interest. Here, we assessed the association among enterobacterial flora, dietary factors, and ulcerative colitis (UC) progression. Forty-six patients with UC who were diagnosed as being in remission were enrolled. We collected each patient's stool sample one or two days before diagnostic colonoscopy. After colonoscopy, we observed the patients for one year and then retrospectively divided them into two groups: remission (n = 39) and relapse (n = 7) groups, depending on whether the relapse occurred during the follow-up period, and analyzed the relationship among patient characteristics, dietary factors, enterobacterial flora, and UC relapse. Overall, there were no significant differences in bacterial community populations between the remission and relapse groups, except that the order Lactobacillales was detected at a significantly higher rate in the relapse than in the remission group (100% vs 71.4%, p<0.05). Vitamin C intake was significantly higher in the remission than in the relapse group (p<0.05). Although there were no obvious differences in enterobacterial flora between the remission and relapse groups, there was a relationship among enterobacterial flora, diet, and UC progression. Given that the enterobacterial flora was only analyzed at the initiation of the study, we conclude that in future analyses, enterobacterial flora should be sampled at numerous time points to examine its role in UC progression. Further long-term longitudinal studies examining enterobacterial flora, dietary factors, and UC progression are also required.
Collapse
Affiliation(s)
- Tomoya Sugiyama
- Department of Gastroenterology, Aichi Medical University School of Medicine, 1-1 Yazakokarimata, Nagakute, Aichi 480-1195, Japan
| | - Makoto Sasaki
- Department of Gastroenterology, Aichi Medical University School of Medicine, 1-1 Yazakokarimata, Nagakute, Aichi 480-1195, Japan
| | - Shoko Nakagawa
- Department of Gastroenterology, Aichi Medical University School of Medicine, 1-1 Yazakokarimata, Nagakute, Aichi 480-1195, Japan
| | - Satoshi Inoue
- Department of Gastroenterology, Aichi Medical University School of Medicine, 1-1 Yazakokarimata, Nagakute, Aichi 480-1195, Japan
| | - Kazunori Adachi
- Department of Gastroenterology, Aichi Medical University School of Medicine, 1-1 Yazakokarimata, Nagakute, Aichi 480-1195, Japan
| | - Takashi Yoshimine
- Department of Gastroenterology, Aichi Medical University School of Medicine, 1-1 Yazakokarimata, Nagakute, Aichi 480-1195, Japan
| | - Yoshiharu Yamaguchi
- Department of Gastroenterology, Aichi Medical University School of Medicine, 1-1 Yazakokarimata, Nagakute, Aichi 480-1195, Japan
| | - Yasuhiro Tamura
- Department of Gastroenterology, Aichi Medical University School of Medicine, 1-1 Yazakokarimata, Nagakute, Aichi 480-1195, Japan
| | - Shinya Izawa
- Department of Gastroenterology, Aichi Medical University School of Medicine, 1-1 Yazakokarimata, Nagakute, Aichi 480-1195, Japan
| | - Yasutaka Hijikata
- Department of Gastroenterology, Aichi Medical University School of Medicine, 1-1 Yazakokarimata, Nagakute, Aichi 480-1195, Japan
| | - Masahide Ebi
- Department of Gastroenterology, Aichi Medical University School of Medicine, 1-1 Yazakokarimata, Nagakute, Aichi 480-1195, Japan
| | - Mari Mizuno
- Department of Gastroenterology, Aichi Medical University School of Medicine, 1-1 Yazakokarimata, Nagakute, Aichi 480-1195, Japan
| | - Sayuri Yamamoto
- Department of Gastroenterology, Aichi Medical University School of Medicine, 1-1 Yazakokarimata, Nagakute, Aichi 480-1195, Japan
| | - Yasushi Funaki
- Department of Gastroenterology, Aichi Medical University School of Medicine, 1-1 Yazakokarimata, Nagakute, Aichi 480-1195, Japan
| | - Naotaka Ogasawara
- Department of Gastroenterology, Aichi Medical University School of Medicine, 1-1 Yazakokarimata, Nagakute, Aichi 480-1195, Japan
| | - Chiho Goto
- Department of Health and Nutrition, Faculty of Health and Human Life, Nagoya Bunri University, 365 Maeda, Inazawa-city, Inazawa, Aichi 492-8520, Japan
| | - Kunio Kasugai
- Department of Gastroenterology, Aichi Medical University School of Medicine, 1-1 Yazakokarimata, Nagakute, Aichi 480-1195, Japan
| |
Collapse
|
69
|
Kridtayopas C, Rakangtong C, Bunchasak C, Loongyai W. Effect of prebiotic and synbiotic supplementation in diet on growth performance, small intestinal morphology, stress, and bacterial population under high stocking density condition of broiler chickens. Poult Sci 2019; 98:4595-4605. [PMID: 30951594 DOI: 10.3382/ps/pez152] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 03/11/2019] [Indexed: 01/05/2023] Open
Abstract
The current study investigated the effect of prebiotic mannan-oligosaccharide (MOS) and synbiotic (MOS mixed with Bacillus subtilis and Bacillus licheniformis) on growth performance and bacterial population under high stocking density (HSD) conditions in broilers. A total of 605 one-day-old male Arbor Acres broiler chickens were randomly assigned to 4 treatments: normal stocking density (NSD; 30 kg/m2 fed basal diets), HSD (40 kg/m2 fed basal diets), HSD chickens fed 0.1% prebiotic (HSDp), and HSD fed 0.1% synbiotic (HSDs). At 35 D of age, the body weight of HSD and HSDp were poorer than NSD group (P < 0.01), whereas the feed conversion ratio (FCR) of the HSDs) group was better than the NSD group (P < 0.01). The HSDp and HSDs groups improved FCR (P < 0.01) and has cheaper feed cost per gain compared to the HSD group. Moreover, the body weight of HSDs group was heavier than the HSDp group (P < 0.05). The level of corticosterone and the heterophil to lymphocyte ratio were highest in the HSD group, whereas these indexes were reduced in both HSDp and HSDs groups (P < 0.05). Duodenal, jejunal, and ileal villus heights were shortest in the HSD group (P < 0.01), and the lowest ileal segment goblet cell counts were also observed in this group (P < 0.05). The HSDp and HSDs groups improved the morphology of gastrointestinal (GI) tract (P < 0.05). The Lactobacillus sp. and Clostridium sp. count in the GI tract of HSD group were low (P < 0.01), whereas Escherichia coli was high (P < 0.01), and Salmonella spp. in jejunum and cecum were detectable when compared with NSD group. Conversely, Bacillus sp., Lactobacillus sp., and Clostridium sp. in HSDp and HSDs groups were increased, and E. coli was reduced in the HSDs group (P < 0.01). Therefore, it is clear that stress from HSD negatively affected growth performance, gut morphology, and microbial population, whereas the supplementation of prebiotic or synbiotic can mitigate the effect of stress and microbial dysbiosis in gut of broiler chickens under HSD condition. Comparatively, under this condition, using synbiotic appears to have more beneficial effects than using the prebiotic.
Collapse
Affiliation(s)
- Chayatid Kridtayopas
- Department of Animal Sciences, Faculty of Agriculture, Kasetsart University, 10900 Bangkok, Thailand
| | - Choawit Rakangtong
- Department of Animal Sciences, Faculty of Agriculture, Kasetsart University, 10900 Bangkok, Thailand
| | - Chaiyapoom Bunchasak
- Department of Animal Sciences, Faculty of Agriculture, Kasetsart University, 10900 Bangkok, Thailand
| | - Wiriya Loongyai
- Department of Animal Sciences, Faculty of Agriculture, Kasetsart University, 10900 Bangkok, Thailand
| |
Collapse
|
70
|
Wu KQ, Sun WJ, Li N, Chen YQ, Wei YL, Chen DF. Small intestinal bacterial overgrowth is associated with Diarrhea-predominant irritable bowel syndrome by increasing mainly Prevotella abundance. Scand J Gastroenterol 2019; 54:1419-1425. [PMID: 31765575 DOI: 10.1080/00365521.2019.1694067] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Objective: Diarrhea-predominant irritable bowel syndrome (IBS-D) is the main subtype of IBS, a chronic functional gastrointestinal disorder. Small intestinal bacterial overgrowth (SIBO), which is characterized by dysbiosis of the bowel, causes gastrointestinal symptoms quite similar to IBS-D. However, whether SIBO correlates with IBS-D and its further mechanism remain unknown.Materials and Methods: The study included 60 IBS-D patients that fulfilled Rome IV criteria and 60 healthy controls. All subjects were undergoing a lactose breath test (LBT) to diagnose SIBO. IBS-D patients were further assigned to negative SIBO (SIBO-) subgroup and positive SIBO (SIBO+) subgroup to analyze the scores of symptoms and differences in the fecal microbiota.Results: The prevalence of SIBO in IBS-D patients was higher than that in healthy controls (51.7% vs. 16.7%, p ≤ .001). In addition, IBS-SSS in SIBO+ subgroup was significantly higher than SIBO- subgroup (p = .015). The 16S rRNA analyses showed that composition and abundance of fecal microbiota were obviously different between the two subgroups. There was a remarkable increase in Prevotella in IBS-D patients, especially in IBS-D SIBO+ sufferers. Meanwhile, there were a moderately positive correlation of the abundance of Prevotella (rho = 0.458, p ≤ .001) with IBS-SSS.Conclusion: SIBO is associated with IBS-D, which may be related to alteration in the intestinal microbiota. These findings suggest the potent role of Prevotella in gastrointestinal symptoms between SIBO and IBS-D, thus provide a novel insight into the connection between SIBO and IBS-D.
Collapse
Affiliation(s)
- Kang-Qi Wu
- Department of Gastroenterology, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, People's Republic of China
| | - Wen-Jing Sun
- Department of Gastroenterology, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, People's Republic of China
| | - Ning Li
- Department of Gastroenterology, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, People's Republic of China
| | - Yu-Qin Chen
- Department of Gastroenterology, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, People's Republic of China
| | - Yan-Ling Wei
- Department of Gastroenterology, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, People's Republic of China
| | - Dong-Feng Chen
- Department of Gastroenterology, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, People's Republic of China
| |
Collapse
|
71
|
Gao Z, Chen KY, Mueller O, Zhang H, Rakhilin N, Chen J, Shen X. Microbiota of Inflammatory Bowel Disease Models. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2019; 2018:2374-2377. [PMID: 30440884 DOI: 10.1109/embc.2018.8512848] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Gut microbiome plays an important role in inflammatory bowel disease (IBD), a group of intestinal chronic inflammation conditions that affect a large population. The animal models of IBD have long been established on basis of pathological features, but their ability to recapitulate patient gut microbiota is unknown. We investigated and compared the composition and biodiversity of bacterial population in the fecal samples from rat models of the two IBD subtypes, and compared them with patient samples. Our analyses revealed that inflammation reduces overall microbiome diversity and increased variation between individuals. We identified specific microbial signatures associated with the two IBD subtypes that were consistent between the animal models and human IBD patients, suggesting that the animal models can partially recapitulate the microbiota in human diseases. Furthermore, metagenome prediction analysis suggested microbial functions that were likely altered by host-microbiota interactions in IBD models.
Collapse
|
72
|
Palacios-González B, Meraz-Cruz N, Valdez-Palomares F, Nambo-Venegas R. Equibiotic-GI Consumption Improves Intestinal Microbiota in Subjects with Functional Dyspepsia. CURRENT DRUG THERAPY 2019. [DOI: 10.2174/1574885514666190212114412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:At present, the interpretation of any dysfunction by pathogenic microbial colonization of the digestive tract can be considered as the rupture of the microbiotic balance in the injured or infected area. Phytodrugs with useful properties to balance the intestinal microbiota equibiotics represent an alternative recently proposed by the Medicinal Plant Research Company Phytomedicamenta S.A. The Equibiotic-GI® is a phytodrug developed as a combination of two plant extracts, obtained from the leaves of Psidium guajava L, (Myrtaceae) and the roots of Coptis chinensis Franch. (Racunculaceae). Both plants used traditionally for the treatment of several gastrointestinal disorders.Objective:The aim of the current study was to assess the effect of Equibiotic-GI® suspension on intestinal microbiota of subjects with functional dyspepsia.Methods:An open-label study performed in 8 adult subjects with functional dyspepsia receiving orally 20 mL of the suspension, daily for two weeks. Fecal samples were collected at baseline and the end of treatment for assessing gut microbiota composition by sequencing the V3-V4 region of the 16S rRNA gene.Results:Equibiotic-GI modified the Bacteriodetes/Firmicutes proportion increasing the richness of the microbiota composition and Rikenellaceae and Alistipes abundance.Conclusion:Together with the improvement in the gastrointestinal symptomatology after the consumption of the product, the present study is the first clinical demonstration of the capacity of the Equibiotic-GI® to restore and balance the intestinal microbiota.
Collapse
Affiliation(s)
- Berenice Palacios-González
- Unidad de Vinculación Científica de la Facultad de Medicina UNAM-Instituto Nacional de Medicina Genómica (INMEGEN), México City, Mexico
| | - Noemí Meraz-Cruz
- Unidad de Vinculación Científica de la Facultad de Medicina UNAM-Instituto Nacional de Medicina Genómica (INMEGEN), México City, Mexico
| | - Fernanda Valdez-Palomares
- Unidad de Vinculación Científica de la Facultad de Medicina UNAM-Instituto Nacional de Medicina Genómica (INMEGEN), México City, Mexico
| | - Rafael Nambo-Venegas
- Laboratorio Bioquímica de Enfermedades Crónicas, Instituto Nacional de Medicina Genómica (INMEGEN), México City, Mexico
| |
Collapse
|
73
|
Zhong H, Ren H, Lu Y, Fang C, Hou G, Yang Z, Chen B, Yang F, Zhao Y, Shi Z, Zhou B, Wu J, Zou H, Zi J, Chen J, Bao X, Hu Y, Gao Y, Zhang J, Xu X, Hou Y, Yang H, Wang J, Liu S, Jia H, Madsen L, Brix S, Kristiansen K, Liu F, Li J. Distinct gut metagenomics and metaproteomics signatures in prediabetics and treatment-naïve type 2 diabetics. EBioMedicine 2019; 47:373-383. [PMID: 31492563 PMCID: PMC6796533 DOI: 10.1016/j.ebiom.2019.08.048] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 08/19/2019] [Accepted: 08/22/2019] [Indexed: 12/12/2022] Open
Abstract
Background The gut microbiota plays important roles in modulating host metabolism. Previous studies have demonstrated differences in the gut microbiome of T2D and prediabetic individuals compared to healthy individuals, with distinct disease-related microbial profiles being reported in groups of different age and ethnicity. However, confounding factors such as anti-diabetic medication hamper identification of the gut microbial changes in disease development. Method We used a combination of in-depth metagenomics and metaproteomics analyses of faecal samples from treatment-naïve type 2 diabetic (TN-T2D, n = 77), pre-diabetic (Pre-DM, n = 80), and normal glucose tolerant (NGT, n = 97) individuals to investigate compositional and functional changes of the gut microbiota and the faecal content of microbial and host proteins in Pre-DM and treatment-naïve T2D individuals to elucidate possible host-microbial interplays characterizing different disease stages. Findings We observed distinct differences characterizing the gut microbiota of these three groups and validated several key features in an independent TN-T2D cohort. We also demonstrated that the content of several human antimicrobial peptides and pancreatic enzymes differed in faecal samples between three groups. Interpretation Our findings suggest a complex, disease stage-dependent interplay between the gut microbiota and the host and point to the value of metaproteomics to gain further insight into interplays between the gut microbiota and the host. Fund The study was supported by the National Natural Science Foundation of China (No. 31601073), the National Key Research and Development Program of China (No. 2017YFC0909703) and the Shenzhen Municipal Government of China (No. JCYJ20170817145809215). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
Collapse
Affiliation(s)
- Huanzi Zhong
- BGI-Shenzhen, Shenzhen 518083, China; China National GeneBank, Shenzhen 518120, China; Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Huahui Ren
- BGI-Shenzhen, Shenzhen 518083, China; China National GeneBank, Shenzhen 518120, China; Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Yan Lu
- Suzhou Centre for Disease Control and Prevention, Suzhou 215007, China
| | - Chao Fang
- BGI-Shenzhen, Shenzhen 518083, China; China National GeneBank, Shenzhen 518120, China; Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Guixue Hou
- BGI-Shenzhen, Shenzhen 518083, China; China National GeneBank, Shenzhen 518120, China
| | - Ziyi Yang
- BGI-Shenzhen, Shenzhen 518083, China; China National GeneBank, Shenzhen 518120, China
| | - Bing Chen
- BGI-Shenzhen, Shenzhen 518083, China; China National GeneBank, Shenzhen 518120, China
| | - Fangming Yang
- BGI-Shenzhen, Shenzhen 518083, China; BGI Education Centre, University of Chinese Academy of Sciences, Shenzhen 518083, China
| | - Yue Zhao
- BGI-Shenzhen, Shenzhen 518083, China; China National GeneBank, Shenzhen 518120, China
| | - Zhun Shi
- BGI-Shenzhen, Shenzhen 518083, China; China National GeneBank, Shenzhen 518120, China
| | - Baojin Zhou
- BGI-Shenzhen, Shenzhen 518083, China; China National GeneBank, Shenzhen 518120, China
| | - Jiegen Wu
- BGI-Shenzhen, Shenzhen 518083, China
| | - Hua Zou
- BGI-Shenzhen, Shenzhen 518083, China; BGI Education Centre, University of Chinese Academy of Sciences, Shenzhen 518083, China
| | - Jin Zi
- BGI-Shenzhen, Shenzhen 518083, China; China National GeneBank, Shenzhen 518120, China
| | - Jiayu Chen
- China National GeneBank, Shenzhen 518120, China
| | - Xiao Bao
- China National GeneBank, Shenzhen 518120, China
| | - Yihe Hu
- Suzhou Centre for Disease Control and Prevention, Suzhou 215007, China
| | - Yan Gao
- Suzhou Centre for Disease Control and Prevention, Suzhou 215007, China
| | - Jun Zhang
- Suzhou Centre for Disease Control and Prevention, Suzhou 215007, China
| | - Xun Xu
- BGI-Shenzhen, Shenzhen 518083, China; China National GeneBank, Shenzhen 518120, China
| | - Yong Hou
- BGI-Shenzhen, Shenzhen 518083, China; China National GeneBank, Shenzhen 518120, China
| | - Huanming Yang
- BGI-Shenzhen, Shenzhen 518083, China; James D. Watson Institute of Genome Sciences, Hangzhou 310058, China
| | - Jian Wang
- BGI-Shenzhen, Shenzhen 518083, China; James D. Watson Institute of Genome Sciences, Hangzhou 310058, China
| | - Siqi Liu
- BGI-Shenzhen, Shenzhen 518083, China; China National GeneBank, Shenzhen 518120, China
| | - Huijue Jia
- BGI-Shenzhen, Shenzhen 518083, China; China National GeneBank, Shenzhen 518120, China
| | - Lise Madsen
- BGI-Shenzhen, Shenzhen 518083, China; China National GeneBank, Shenzhen 518120, China; Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, 2100 Copenhagen, Denmark; Institute of Marine Research, P.O. Box 7800, 5020 Bergen, Norway
| | - Susanne Brix
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Soltofts Plads, 2800 Kgs. Lyngby, Denmark
| | - Karsten Kristiansen
- BGI-Shenzhen, Shenzhen 518083, China; China National GeneBank, Shenzhen 518120, China; Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, 2100 Copenhagen, Denmark.
| | - Fang Liu
- Suzhou Centre for Disease Control and Prevention, Suzhou 215007, China.
| | - Junhua Li
- BGI-Shenzhen, Shenzhen 518083, China; China National GeneBank, Shenzhen 518120, China; School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China.
| |
Collapse
|
74
|
Ballini A, Santacroce L, Cantore S, Bottalico L, Dipalma G, Topi S, Saini R, De Vito D, Inchingolo F. Probiotics Efficacy on Oxidative Stress Values in Inflammatory Bowel Disease: A Randomized Double-Blinded Placebo-Controlled Pilot Study. Endocr Metab Immune Disord Drug Targets 2019; 19:373-381. [PMID: 30574857 DOI: 10.2174/1871530319666181221150352] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 11/18/2018] [Accepted: 11/28/2018] [Indexed: 01/14/2023]
Abstract
BACKGROUND Inflammatory bowel diseases (IBD), which include Crohn's disease (CD) and ulcerative colitis (UC), are described as a chronic inflammation of the small intestine and colon, caused by a dysregulated immune response to host intestinal microbiota in genetically susceptible subjects. OBJECTIVE The aim of this study was to compare probiotic therapy versus placebo in Oxidative Stress Values and clinical features in patients affected by IBD. METHOD Forty (40) patients previously diagnosed for IBD were recruited and randomized to receive probiotics (test group, n=20) or placebo (control group, n=20) administered for 90 days. Subjects in both the groups were assessed for overall oxidant ability (d-ROMs test) and for the antioxidant response (BAP test): data were reported at baseline, after 1 and 3 months. Additional data from anamnesis and haematological investigation were also reported during the study. RESULTS d-ROM assay clearly showed that the values observed in the test group were significantly improved, leading to oxidative stress values which are not pathological. The test group showed increasing BAP values, thus confirming the overall improvements of patients 'health following administration of probiotics. CONCLUSION Oral administration of the specific probiotics demonstrated its efficacy and safety on patients affected by IBD.
Collapse
Affiliation(s)
- Andrea Ballini
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari "Aldo Moro", Bari, Italy.,Interdisciplinary Department of Medicine, University of Bari "Aldo Moro", Bari, Italy
| | - Luigi Santacroce
- Ionian Department, University of Bari "Aldo Moro", Bari, Italy.,Polypheno - Academic Spin Off, University of Bari "Aldo Moro", Bari, Italy.,School of Technical Medical Sciences, "A. Xhuvani" University, Elbasan, Albania
| | - Stefania Cantore
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari "Aldo Moro", Bari, Italy.,Interdisciplinary Department of Medicine, University of Bari "Aldo Moro", Bari, Italy
| | - Lucrezia Bottalico
- Polypheno - Academic Spin Off, University of Bari "Aldo Moro", Bari, Italy
| | - Gianna Dipalma
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari "Aldo Moro", Bari, Italy.,Interdisciplinary Department of Medicine, University of Bari "Aldo Moro", Bari, Italy
| | - Skender Topi
- Ionian Department, University of Bari "Aldo Moro", Bari, Italy.,Polypheno - Academic Spin Off, University of Bari "Aldo Moro", Bari, Italy.,School of Technical Medical Sciences, "A. Xhuvani" University, Elbasan, Albania
| | - Rajiv Saini
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari "Aldo Moro", Bari, Italy
| | - Danila De Vito
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari "Aldo Moro", Bari, Italy
| | - Francesco Inchingolo
- Interdisciplinary Department of Medicine, University of Bari "Aldo Moro", Bari, Italy
| |
Collapse
|
75
|
Schreiner P, Neurath MF, Ng SC, El-Omar EM, Sharara AI, Kobayashi T, Hisamatsu T, Hibi T, Rogler G. Mechanism-Based Treatment Strategies for IBD: Cytokines, Cell Adhesion Molecules, JAK Inhibitors, Gut Flora, and More. Inflamm Intest Dis 2019; 4:79-96. [PMID: 31559260 DOI: 10.1159/000500721] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Accepted: 05/02/2019] [Indexed: 12/14/2022] Open
Abstract
Background Although TNF inhibitors revolutionized the therapy of inflammatory bowel disease (IBD), we have been reaching a point where other therapies with different mechanisms of action are necessary. A rising number of elderly IBD patients with contraindications to established therapies and a growing group of patients losing response to anti-TNF therapy compel us to find safer, better-tolerated, and, ideally, personalized treatment options. However, in order to choose the right drug to fit a patient, it is indispensable to understand the pathomechanism involved in IBD. Summary The aim of this review is to explain the inflammatory signaling pathways in IBD and how to inhibit them with current and future therapeutic approaches. Next to biologic agents targeting inflammatory cytokines (anti-TNF agents, anti-IL-12/-23 agents, and specific inhibitors of IL-23), biologics blocking leukocyte trafficking to the gut (anti-integrin antibodies) are available nowadays. More recently, small molecules inhibiting the JAK-STAT pathway (JAK inhibitors) or preventing lymphocyte trafficking (sphingosine-1-phosphate modulators) have been approved or are under investigation. Furthermore, modifying the microbiota has potential therapeutic effects on IBD, and autologous hematopoietic or mesenchymal stem cell transplantation may be considered for a highly selected group of IBD patients. Key Message Physicians should understand the different mechanisms of action of the potential therapies for IBD to select the right drug for the right patient.
Collapse
Affiliation(s)
- Philipp Schreiner
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Markus F Neurath
- Medizinische Klinik 1, Universitätsklinikum Erlangen-Nürnberg, Erlangen, Germany
| | - Siew C Ng
- Department of Medicine and Therapeutics, Institute of Digestive Disease, LKS Institute of Health Science, State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
| | - Emad M El-Omar
- St. George and Sutherland Clinical School, University of New South Wales, Sydney, New South Wales, Australia
| | - Ala I Sharara
- Division of Gastroenterology, Department of Internal Medicine, American University of Beirut, Beirut, Lebanon
| | - Taku Kobayashi
- Center for Advanced IBD Research and Treatment, Kitasato University Kitasato Institute Hospital, Tokyo, Japan
| | | | - Toshifumi Hibi
- Center for Advanced IBD Research and Treatment, Kitasato University Kitasato Institute Hospital, Tokyo, Japan
| | - Gerhard Rogler
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
76
|
Ayers TD, Leonard-Puppa E, Kader HA, Waddell J, Watkins RD, Blanchard SS, Safta AM, Rawal N. Oral Vancomycin as an Adjuvant Treatment in IBD. CROHN'S & COLITIS 360 2019. [DOI: 10.1093/crocol/otz015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Abstract
Objective
To study the efficacy of oral vancomycin (POV) treatment in pediatric inflammatory bowel disease (IBD).
Methods
We conducted retrospective and prospective chart reviews, identifying patients using the Division’s Inflammatory Bowel Disease (IBD) registry, ICD-9 and ICD-10 codes for IBD, and recall of patients receiving POV. Patients aged 2–21 years with active IBD at initiation of POV were included unless they had Clostridium difficile infection or primary sclerosing cholangitis (PSC). Pre- and posttreatment analysis included a Physician Global Assessment (PGA), pediatric ulcerative colitis (UC) activity index (PUCAI), and an abbreviated pediatric Crohn’s disease (CD) activity index (PCDAI). The Wilcoxon Signed Ranks test, determined if pre- and post-POV rankings of symptom severity differed. Mann–Whitney U tests assessed improvement in presenting symptoms.
Results
Nineteen patients met inclusion criteria (12 CD and 7 UC). POV improved the PGA score in 16 of 19 patients (P < 0.001). Mean PGA score pretreatment was 3 ± 0.471; posttreatment mean of 1.58 ± 0.769. Abdominal pain (P < 0.001), diarrhea (P < 0.002), anemia (P < 0.002), and blood in stool (P < 0.001) showed significant improvement. PUCAI and PCDAI scores, pretreatment means of 50 ± 17 and 33 ± 9, respectively, also improved with mean score reduction of 23 in CD and 38 in UC patients after POV initiation (P-value < 0.0001). This improvement was noted for both IBD subtypes.
Conclusions
POV may be an effective adjuvant treatment for pediatric IBD. Its effectiveness is likely due to a combination of its anti-tumor necrosis factor alpha-α activity and its influence on the gut microbiome. Further controlled studies of POV in IBD are warranted to determine the most efficacious use of POV in pediatric IBD.
Aim
This study attempts to expand on the current literature to determine efficacy of POV as an adjuvant therapy in treating active IBD in children.
Collapse
Affiliation(s)
- Travis D Ayers
- Pediatric GI, University of Arkansas for Medical Sciences, Arkansas Children’s Hospital, Little Rock, AR
| | | | - Howard A Kader
- Pediatric GI, University of Maryland School of Medicine, Baltimore, MD
| | - Jaylyn Waddell
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD
| | - Runa D Watkins
- Pediatric GI, University of Maryland School of Medicine, Baltimore, MD
| | - Samra S Blanchard
- Pediatric GI, University of Maryland School of Medicine, Baltimore, MD
| | - Anca M Safta
- Pediatric GI, Wake Forest School of Medicine, Winston-Salem, NC
| | - Nidhi Rawal
- Pediatric GI, University of Maryland School of Medicine, Baltimore, MD
| |
Collapse
|
77
|
Pan LL, Niu W, Fang X, Liang W, Li H, Chen W, Zhang H, Bhatia M, Sun J. Clostridium butyricum Strains Suppress Experimental Acute Pancreatitis by Maintaining Intestinal Homeostasis. Mol Nutr Food Res 2019; 63:e1801419. [PMID: 31034143 DOI: 10.1002/mnfr.201801419] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 04/13/2019] [Indexed: 01/24/2023]
Abstract
SCOPE Acute pancreatitis (AP) is a common abdominal inflammatory disease. Disturbed gut homeostasis secondary to pancreatic inflammation aggravates the condition retroactively. The current study investigates potential beneficial effects of Clostridium butyricum (C. butyricum) strains on AP and underlying mechanisms. METHODS AND RESULTS C. butyricum strains MIYAIRI 588 (CBM588) and CGMCC0313.1 (CB0313.1) were supplemented to mice for three weeks before experimental AP or SAP induction. Both CBM588 and CB0313.1 protected against AP, as evidenced by reduced serum amylase and lipase levels, pancreatic edema, and myeloperoxidase activity. Amelioration of both experimental AP and SAP by CB0313.1 indicated a non-model-specific effect. Moreover, C. butyricum inhibited pancreatic neutrophil and dendritic cell infiltration, nucleotide-binding domain leucine-rich repeat-containing family, pyrin domain-containing 3 inflammasome activation, and pro-inflammatory pathways. Additionally in the gut, C. butyricum strains attenuated AP-associated intestinal inflammation and barrier dysfunction, accompanied with reduced pathogenic bacteria Escherichia coli and Enterococcus penetration into pancreas. Gut microbiome analyses further revealed that beneficial effects of C. butyricum on pancreatic-gut homeostasis were correlated with improved dysbiosis. In particular, relative abundance of Desulfovibrionaceae decreased, and Verrucomicrobiaceae Clostridiaceae and Lactobacillaceae increased. CONCLUSIONS For the first time, a protective effect of C. butyricum in AP by modulating intestinal homeostasis is demonstrated.
Collapse
Affiliation(s)
- Li-Long Pan
- School of Medicine, Jiangnan University, Wuxi, 214122, P. R. China
| | - Wenying Niu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China.,School of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China
| | - Xin Fang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China.,School of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China
| | - Wenjie Liang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China.,School of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China
| | - Hongli Li
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China.,School of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China.,School of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China.,School of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China
| | - Madhav Bhatia
- Inflammation Research Group, Department of Pathology, University of Otago, Christchurch, 8140, New Zealand
| | - Jia Sun
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China.,School of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China
| |
Collapse
|
78
|
Jo SH, Park HG, Song WS, Kim SM, Kim EJ, Yang YH, Kim JS, Kim BG, Kim YG. Structural characterization of phosphoethanolamine-modified lipid A from probiotic Escherichia coli strain Nissle 1917. RSC Adv 2019; 9:19762-19771. [PMID: 35519361 PMCID: PMC9065436 DOI: 10.1039/c9ra02375e] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 06/19/2019] [Indexed: 01/05/2023] Open
Abstract
Gut microbiota, a complex microbial community inhabiting human or animal intestines recently regarded as an endocrine organ, has a significant impact on human health. Probiotics can modulate gut microbiota and the gut environment by releasing a range of bioactive compounds. Escherichia coli (E. coli) strain Nissle 1917 (EcN), a Gram-negative bacterial strain, has been used to treat gastrointestinal (GI) disorders (i.e., inflammatory bowel disease, diarrhea, ulcerative colitis, and so on). However, endotoxicity of lipopolysaccharide (LPS), a major component of the cell wall of Gram-negative bacteria in the gut, is known to have a strong influence on gut inflammation and maintenance of gut homeostasis. Therefore, characterizing the chemical structure of lipid A which determines the toxicity of LPS is needed to understand nonpathogenic colonization and commensalism properties of EcN in the gut more precisely. In the present study, MALDI multiple-stage mass spectrometry analysis of lipid A extracted from EcN demonstrates that hexaacylated lipid A (m/z 1919.19) contains a glucosamine disaccharide backbone, a myristate, a laurate, four 3-hydroxylmyristates, two phosphates, and phosphoethanolamine (PEA). PEA modification of lipid A is known to contribute to cationic antimicrobial peptide (CAMP) resistance of Gram-negative bacteria. To confirm the role of PEA in CAMP resistance of EcN, minimum inhibitory concentrations (MICs) of polymyxin B and colistin were determined using a wild-type strain and a mutant strain with deletion of eptA gene encoding PEA transferase. Our results confirmed that MICs of polymyxin B and colistin for the wild-type were twice as high as those for the mutant. These results indicate that EcN can more efficiently colonize the intestine through PEA-mediated tolerance despite the presence of CAMPs in human gut such as human defensins. Thus, EcN can be used to help treat and prevent many GI disorders.
Collapse
Affiliation(s)
- Sung-Hyun Jo
- Department of Chemical Engineering, Soongsil University 369 Sangdo-Ro Seoul 06978 Korea +82-2-828-7099
| | - Han-Gyu Park
- Department of Chemical Engineering, Soongsil University 369 Sangdo-Ro Seoul 06978 Korea +82-2-828-7099
| | - Won-Suk Song
- School of Chemical and Biological Engineering, Seoul National University Seoul 08826 Korea
| | - Seong-Min Kim
- Department of Chemical Engineering, Soongsil University 369 Sangdo-Ro Seoul 06978 Korea +82-2-828-7099
| | - Eun-Jung Kim
- Institute of Molecular Biology and Genetics, Seoul National University Seoul 08826 Korea
| | - Yung-Hun Yang
- Department of Biological Engineering, Konkuk University Seoul 05029 Korea
| | - Jae-Seok Kim
- Department of Laboratory Medicine, Kangdong Sacred Heart Hospital, Hallym University College of Medicine Seoul 05355 Korea
| | - Byung-Gee Kim
- School of Chemical and Biological Engineering, Seoul National University Seoul 08826 Korea
| | - Yun-Gon Kim
- Department of Chemical Engineering, Soongsil University 369 Sangdo-Ro Seoul 06978 Korea +82-2-828-7099
| |
Collapse
|
79
|
Wang L, Tang H, Wang C, Hu Y, Wang S, Shen L. Aquaporin 4 deficiency alleviates experimental colitis in mice. FASEB J 2019; 33:8935-8944. [PMID: 31034776 DOI: 10.1096/fj.201802769rr] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Aquaporin (AQP) 4 is expressed in the basolateral membrane of colonic epithelial cells, and the purpose of this study was to explore the mechanistic role of AQP4 in experimental colitis. Experimental colitis was induced in AQP4 knockout (AQP4-/-) CD-1 mice and AQP4 wild-type (AQP4wt) mice by oral administration of dextran sulfate sodium (DSS). Experimental colitis was clinically established. Compared with AQP4wt mice, AQP4-/- mice showed increased tolerance to DSS-induced experimental colitis, including lesser degree of weight loss, diarrhea and bleeding, lower disease activity index scores, longer colon lengths, and lesser histologic scores. DSS-treated AQP4-/- mice had lower serum levels of IL-6 and TNF, higher IL-10 level, and lesser inflammatory cell infiltration. DSS-treated AQP4-/- mice also had lower immunostaining of NF-κB p65 as well as nuclear levels of p65 and phosphorylated p65. Sequencing of 16S rRNA indicated that DSS-treated AQP4-/- mice maintained intestinal microbial diversity and had higher Firmicutes/Bacteroidetes ratios and greater relative abundance of Erysipelotrichaceae species. These results suggested for the first time that AQP4 deficiency alleviates experimental colitis in mice. Our study helps to understand the pathogenesis of inflammatory bowel diseases, and blocking AQP4 may represent a novel therapeutic approach for ulcerative colitis.-Wang, L., Tang, H., Wang, C., Hu, Y., Wang, S., Shen, L. Aquaporin 4 deficiency alleviates experimental colitis in mice.
Collapse
Affiliation(s)
- Liuhua Wang
- Division of Gastrointestinal Surgery, Department of General Surgery, First Affiliated Hospital, Nanjing Medical University, Nanjing, China.,Department of General Surgery, Northern Jiangsu People's Hospital, Clinical Medical School, Yangzhou University, Yangzhou, China
| | - Hua Tang
- Division of Gastrointestinal Surgery, Department of General Surgery, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Chao Wang
- School of Public Health, Nanjing Medical University, Nanjing, China
| | - Yuhuan Hu
- School of Public Health, Nanjing Medical University, Nanjing, China
| | - Shoulin Wang
- School of Public Health, Nanjing Medical University, Nanjing, China
| | - Lizong Shen
- Division of Gastrointestinal Surgery, Department of General Surgery, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
80
|
Schepper JD, Collins F, Rios-Arce ND, Raehtz S, Schaefer L, Gardinier JD, Britton R, Parameswaran N, McCabe LR. Probiotic Lactobacillus reuteri Prevents Postantibiotic Bone Loss by Reducing Intestinal Dysbiosis and Preventing Barrier Disruption. J Bone Miner Res 2019; 34:681-698. [PMID: 30690795 PMCID: PMC6557403 DOI: 10.1002/jbmr.3635] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 10/24/2018] [Accepted: 11/11/2018] [Indexed: 12/23/2022]
Abstract
Antibiotic treatment, commonly prescribed for bacterial infections, depletes and subsequently causes long-term alterations in intestinal microbiota composition. Knowing the importance of the microbiome in the regulation of bone density, we investigated the effect of postantibiotic treatment on gut and bone health. Intestinal microbiome repopulation at 4-weeks postantibiotic treatment resulted in an increase in the Firmicutes:Bacteroidetes ratio, increased intestinal permeability, and notably reduced femoral trabecular bone volume (approximately 30%, p < 0.01). Treatment with a mucus supplement (a high-molecular-weight polymer, MDY-1001 [MDY]) prevented the postantibiotic-induced barrier break as well as bone loss, indicating a mechanistic link between increased intestinal permeability and bone loss. A link between the microbiome composition and bone density was demonstrated by supplementing the mice with probiotic bacteria. Specifically, Lactobacillus reuteri, but not Lactobacillus rhamnosus GG or nonpathogenic Escherichia coli, reduced the postantibiotic elevation of the Firmicutes:Bacteroidetes ratio and prevented femoral and vertebral trabecular bone loss. Consistent with causing bone loss, postantibiotic-induced dysbiosis decreased osteoblast and increased osteoclast activities, changes that were prevented by both L. reuteri and MDY. These data underscore the importance of microbial dysbiosis in the regulation of intestinal permeability and bone health, as well as identify L. reuteri and MDY as novel therapies for preventing these adverse effects. © 2018 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
| | - Fraser Collins
- Department of Physiology, Michigan State University, East Lansing, Michigan
| | - Naiomy Deliz Rios-Arce
- Department of Physiology, Michigan State University, East Lansing, Michigan
- Comparative Medicine and Integrative Biology Program, Michigan State University, East Lansing, Michigan
| | - Sandi Raehtz
- Department of Physiology, Michigan State University, East Lansing, Michigan
| | - Laura Schaefer
- Department of Molecular Virology and Microbiology, Baylor College of Medicine
| | | | - Robert Britton
- Department of Molecular Virology and Microbiology, Baylor College of Medicine
| | - Narayanan Parameswaran
- Department of Physiology, Michigan State University, East Lansing, Michigan
- equal contribution and co-senior authors
| | - Laura R McCabe
- Department of Physiology, Michigan State University, East Lansing, Michigan
- equal contribution and co-senior authors
| |
Collapse
|
81
|
Mondanelli G, Iacono A, Carvalho A, Orabona C, Volpi C, Pallotta MT, Matino D, Esposito S, Grohmann U. Amino acid metabolism as drug target in autoimmune diseases. Autoimmun Rev 2019; 18:334-348. [DOI: 10.1016/j.autrev.2019.02.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 10/30/2018] [Indexed: 12/14/2022]
|
82
|
Segal JP, Mullish BH, Quraishi MN, Acharjee A, Williams HRT, Iqbal T, Hart AL, Marchesi JR. The application of omics techniques to understand the role of the gut microbiota in inflammatory bowel disease. Therap Adv Gastroenterol 2019; 12:1756284818822250. [PMID: 30719076 PMCID: PMC6348496 DOI: 10.1177/1756284818822250] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 11/23/2018] [Indexed: 02/04/2023] Open
Abstract
The aetiopathogenesis of inflammatory bowel diseases (IBD) involves the complex interaction between a patient's genetic predisposition, environment, gut microbiota and immune system. Currently, however, it is not known if the distinctive perturbations of the gut microbiota that appear to accompany both Crohn's disease and ulcerative colitis are the cause of, or the result of, the intestinal inflammation that characterizes IBD. With the utilization of novel systems biology technologies, we can now begin to understand not only details about compositional changes in the gut microbiota in IBD, but increasingly also the alterations in microbiota function that accompany these. Technologies such as metagenomics, metataxomics, metatranscriptomics, metaproteomics and metabonomics are therefore allowing us a deeper understanding of the role of the microbiota in IBD. Furthermore, the integration of these systems biology technologies through advancing computational and statistical techniques are beginning to understand the microbiome interactions that both contribute to health and diseased states in IBD. This review aims to explore how such systems biology technologies are advancing our understanding of the gut microbiota, and their potential role in delineating the aetiology, development and clinical care of IBD.
Collapse
Affiliation(s)
- Jonathan P. Segal
- Inflammatory Bowel Disease Department, St Mark’s Hospital, Harrow HA1 3UJ, UK
| | - Benjamin H. Mullish
- Division of Integrative Systems Medicine and Digestive Disease, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, UK
| | - Mohammed Nabil Quraishi
- Institute of Immunology and Immunotherapy, University of Birmingham, Department of Gastroenterology, University Hospital, Birmingham, UK
| | - Animesh Acharjee
- College of Medical and Dental Sciences, Institute of Cancer and Genomic Sciences, Centre for Computational Biology, University of Birmingham, Birmingham, UK
- Institute of Translational Medicine, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
- NIHR Surgical Reconstruction and Microbiology Research Centre, Birmingham, UK
| | - Horace R. T. Williams
- Division of Integrative Systems Medicine and Digestive Disease, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, UK
| | - Tariq Iqbal
- Institute of Immunology and Immunotherapy, University of Birmingham, Department of Gastroenterology, University Hospital, Birmingham, UK
| | - Ailsa L. Hart
- Inflammatory Bowel Disease Department, St Mark’s Hospital, Harrow, UK
- Department of Surgery and Cancer, Division of Integrative Systems Medicine and Digestive Disease, Faculty of Medicine, Imperial College, London, UK
| | - Julian R. Marchesi
- Department of Surgery and Cancer, Division of Integrative Systems Medicine and Digestive Disease, Faculty of Medicine, Imperial College, London, UK
- School of Biosciences, Cardiff University, Cardiff, UK
| |
Collapse
|
83
|
Lu J, Dong B, Chen A, He F, Peng B, Wu Z, Cao J, Li W. Escherichia coli promotes DSS‑induced murine colitis recovery through activation of the TLR4/NF‑κB signaling pathway. Mol Med Rep 2019; 19:2021-2028. [PMID: 30664156 PMCID: PMC6390074 DOI: 10.3892/mmr.2019.9848] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 10/12/2018] [Indexed: 12/28/2022] Open
Abstract
Increasing evidence suggests that intestinal microbiota have critical function in the pathogenesis of inflammatory bowel disease. This present study investigated the effects of Escherichia coli (E. coli) in mice with dextran sulfate sodium (DSS)-induced colitis. Furthermore, Toll-like receptor 4 (TLR4) and nuclear factor-κB (NF-κB) gene expression was measured by reverse transcription-quantitative polymerase chain reaction. In total, two experiments were performed. In the first experiment, four groups were established in BALB/c mice: i) Group A, control (no treatments); ii) group B, DSS-induced colitis; iii) group C, DSS-induced colitis bacteria depleted (BD) mice; and iv) group D, E. coli-treated DSS-induced colitis BD mice. In the second experiment, there were three groups: i) Group A1, control C57BL/6 mice; ii) group B1, E. coli-treated DSS-induced colitis BD C57BL/6 mice; and iii) E. coli-treated DSS-induced colitis BD TLR4−/− mice. Clinical outcomes, colon and immune histopathology and tissue myeloperoxidase activity were assessed. Mice with DSS-induced colitis that were treated with E. coli exhibited enhanced recovery, with significantly improved clinical and histological scores compared with the DSS only group. The mRNA expression of TLR4 and NF-κB in the E. coli-treated group was also significantly higher. These effects were abolished in TLR4−/− mice, suggesting that E. coli may have promoted recovery through the TLR4 pathway. The present study indicated that E. coli promoted recovery from DSS-induced colitis in mice, potentially through activation of the TLR4/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Jiabao Lu
- Department of Colorectal Surgery, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510180, P.R. China
| | - Boye Dong
- Department of Colorectal Surgery, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510180, P.R. China
| | - Ailan Chen
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, P.R. China
| | - Feng He
- Department of Colorectal Surgery, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong 510180, P.R. China
| | - Baifu Peng
- Department of Colorectal Surgery, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510180, P.R. China
| | - Zixin Wu
- Department of Colorectal Surgery, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong 510180, P.R. China
| | - Jie Cao
- Department of Colorectal Surgery, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong 510180, P.R. China
| | - Wanglin Li
- Department of Colorectal Surgery, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong 510180, P.R. China
| |
Collapse
|
84
|
Zhou F, Zhu Q, Zheng PF, Feng YL. Association of Fucosyltransferase 2 Gene Variant with Inflammatory Bowel Diseases: A Meta-Analysis. Med Sci Monit 2019; 25:184-192. [PMID: 30615603 PMCID: PMC6338007 DOI: 10.12659/msm.911857] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 09/15/2018] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Ulcerative colitis (UC) and Crohn's disease (CD) are the 2 main type of inflammatory bowel diseases (IBDs). Several studies have been conducted to investigate the association of fucosyltransferase 2 gene (rs601338) variant with UC and CD, but the results were inconsistent. Here, we performed a meta-analysis to clarify this issue based on a relatively larger sample size. MATERIAL AND METHODS A systematic literature search was conducted in PubMed, Embase, CNKI, and Chinese Wangfang databases up to 31 May 2018. Meta results were synthesized by using crude odds ratio with 95% confidence interval. Heterogeneity, sensitivity analysis, subgroup analysis, and publication bias were assessed using STATA 11.0 software. RESULTS A total of 8 relevant studies including 3874 IBDs patients (1872 UC cases, 2002 CD cases) and 5445 controls were included for meta-analysis. We found a significant association between rs601338 A allele and risk of IBDs in the Chinese population (OR=2.35, 95%CI=1.66~3.34, P=0.001), but not in whites. Stratified by disease type, we found a significant association between rs601338 polymorphism with CD and UC in the Chinese population, but not in the white population. In addition, funnel plot and Egger's linear regression test suggests no publication bias in all genetic models. CONCLUSIONS Fucosyltransferase 2 gene (rs601338) polymorphism is associated with susceptibility to IBD, UC, and CD in the Chinese population, but these results might not be generalizable to other ethnic populations. Further well-designed studies are needed to confirm these findings.
Collapse
|
85
|
Sharp RC, Naser ES, Alcedo KP, Qasem A, Abdelli LS, Naser SA. Development of multiplex PCR and multi-color fluorescent in situ hybridization ( m-FISH) coupled protocol for detection and imaging of multi-pathogens involved in inflammatory bowel disease. Gut Pathog 2018; 10:51. [PMID: 30534203 PMCID: PMC6280354 DOI: 10.1186/s13099-018-0278-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 11/29/2018] [Indexed: 12/28/2022] Open
Abstract
Background Several pathogens have been debated to play a role in inflammatory bowel disease (IBD) including Crohn's disease (CD). None of these pathogens have been investigated together in same clinical samples. We developed a multiplex PCR and multi-color fluorescent in situ hybridization (m-FISH) protocols for simultaneous detection of CD-associated pathogens including Mycobacterium avium subspecies paratuberculosis (MAP), Klebsiella pneumoniae, and adherent-invasive Escherichia coli strain LF82. Methods The multiplex PCR is based on 1-h DNAzol® extraction protocol modified for rapid extraction of bacterial DNA from culture, blood, and intestinal biopsies. Oligonucleotide primers sequences unique to these pathogens were evaluated individually and in combinations using bioinformatics and experimental approaches. m-FISH was based on fluorescent-tagged oligonucleotides and confocal scanning laser microscopy (CSLM). Results Following several attempts, the concentration of the oligonucleotide primers and DNA templates and the PCR annealing temperatures were optimized. Multiplex PCR analyses revealed excellent amplification signal in trials where a single primer set and combinations of two and three primers sets were tested against a mixture of DNA from three different bacteria or a mixture of three bacterial cultures mixed in one tube before DNA extraction. Slides with individual and mixtures of bacterial cultures and intestinal tissue sections from IBD patients were tested by m-FISH and the CSLM images verified multiplex PCR results detected on 3% agarose gel. Conclusion We developed a 4-h multiplex PCR protocol, which was validated by m-FISH images, capable of detecting up to four genes from major pathogens associated with CD. The new protocol should serve as an excellent tool to support efforts to study multi-pathogens involved in CD and other autoimmune disease.
Collapse
Affiliation(s)
- Robert C Sharp
- Division of Molecular Microbiology, Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, 4110 Libra Drive, Orlando, FL USA
| | - Ebraheem S Naser
- Division of Molecular Microbiology, Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, 4110 Libra Drive, Orlando, FL USA
| | - Karel P Alcedo
- Division of Molecular Microbiology, Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, 4110 Libra Drive, Orlando, FL USA
| | - Ahmad Qasem
- Division of Molecular Microbiology, Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, 4110 Libra Drive, Orlando, FL USA
| | - Latifa S Abdelli
- Division of Molecular Microbiology, Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, 4110 Libra Drive, Orlando, FL USA
| | - Saleh A Naser
- Division of Molecular Microbiology, Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, 4110 Libra Drive, Orlando, FL USA
| |
Collapse
|
86
|
Effects of the 1975 Japanese diet on the gut microbiota in younger adults. J Nutr Biochem 2018; 64:121-127. [PMID: 30502656 DOI: 10.1016/j.jnutbio.2018.10.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 10/11/2018] [Accepted: 10/29/2018] [Indexed: 12/14/2022]
Abstract
Japan is known for its longevity worldwide; the Japanese diet is thought to contribute to this longevity. However, the Japanese diet has become westernized over the past years, with a parallel increase in the incidence of lifestyle diseases. Thus, whether the modern Japanese diet is still healthy requires investigation. A diet with characteristics of the 1975 Japanese diet (JD) was previously shown to have beneficial effects on mice and humans. In this study, we examined whether intestinal bacteria are involved in the health benefits of this diet by analyzing changes in the composition of the fecal microbiota between humans who ingested the JD and those consuming a modern Japanese diet (MD). We also examined correlations between intestinal bacteria and biological parameters. A randomized controlled trial was performed to determine the effects of the 1975 JD compared to those of the MD. Subjects aged 20-29 years were randomly assigned to the JD (n=11) and MD (n=10) groups. Each subject consumed their respective diet three times per day for 28 days, and changes in intestinal bacteria before to after this period were evaluated. Four genera (unclassified Lachnospiraceae, Parabacteroides, Sutterella and unclassified Rikenellaceae) were significantly changed upon intake of the JD. Based on correlation analysis, relationships were found between changes in these genera and decreases in fat%; fat mass; and levels of blood glutamic oxaloacetic transaminase, blood triacylglycerols and hemoglobin A1c. These results suggest that changes in intestinal bacteria are involved in the health benefits of the JD.
Collapse
|
87
|
Pelgrim CE, Peterson JD, Gosker HR, Schols AMWJ, van Helvoort A, Garssen J, Folkerts G, Kraneveld AD. Psychological co-morbidities in COPD: Targeting systemic inflammation, a benefit for both? Eur J Pharmacol 2018; 842:99-110. [PMID: 30336140 DOI: 10.1016/j.ejphar.2018.10.001] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 09/24/2018] [Accepted: 10/08/2018] [Indexed: 12/15/2022]
Abstract
COPD is a chronic lung disease characterized by persistent respiratory symptoms and airflow limitation due to airway and/or alveolar abnormalities. Furthermore, COPD is often characterized by extrapulmonary manifestations and comorbidities worsening COPD progression and quality of life. A neglected comorbidity in COPD management is mental health impairment defined by anxiety, depression and cognitive problems. This paper summarizes the evidence for impaired mental health in COPD and focuses on current pharmacological intervention strategies. In addition, possible mechanisms in impaired mental health in COPD are discussed with a central role for inflammation. Many comorbidities are associated with multi-organ-associated systemic inflammation in COPD. Considering the accumulative evidence for a major role of systemic inflammation in the development of neurological disorders, it can be hypothesized that COPD-associated systemic inflammation also affects the function of the brain and is an interesting therapeutic target for nutra- and pharmaceuticals.
Collapse
Affiliation(s)
- Charlotte E Pelgrim
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, the Netherlands
| | - Julia D Peterson
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, the Netherlands
| | - Harry R Gosker
- NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Department of Respiratory Medicine, Maastricht, the Netherlands
| | - Annemie M W J Schols
- NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Department of Respiratory Medicine, Maastricht, the Netherlands
| | - Ardy van Helvoort
- NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Department of Respiratory Medicine, Maastricht, the Netherlands; Nutrition, Metabolism and Muscle Sciences, Nutricia Research, Utrecht, the Netherlands
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, the Netherlands; Platform Immunology, Nutricia Research, Utrecht, the Netherlands
| | - Gert Folkerts
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, the Netherlands
| | - Aletta D Kraneveld
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, the Netherlands; Veterinary Pharmacology & Therapeutics, Institute of Risk Assessment Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands.
| |
Collapse
|
88
|
Shinzaki S, Fujii T, Bamba S, Ogawa M, Kobayashi T, Oshita M, Tanaka H, Ozeki K, Takahashi S, Kitamoto H, Kani K, Nanjo S, Sugaya T, Sakakibara Y, Inokuchi T, Kakimoto K, Yamada A, Yasuhara H, Yokoyama Y, Yoshino T, Matsui A, Nakamura M, Tomizawa T, Sakemi R, Kamata N, Hibi T. Seven days triple therapy for eradication of Helicobacter pylori does not alter the disease activity of patients with inflammatory bowel disease. Intest Res 2018; 16:609-618. [PMID: 30301329 PMCID: PMC6223447 DOI: 10.5217/ir.2018.00044] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 06/19/2018] [Indexed: 02/07/2023] Open
Abstract
Background/Aims The influences of Helicobacter pylori eradication therapy on the disease course of inflammatory bowel disease (IBD) are still unclear. We therefore conducted a multicenter, retrospective cohort study to evaluate the safety of H. pylori eradication therapy for IBD patients. Methods IBD patients with H. pylori eradication from 2005 to 2015 (eradication group) and control patients (non-eradication group; 2 paired IBD patients without H. pylori eradication matched with each eradicated patient) were included. IBD exacerbation (increased/additional IBD drug or IBD-associated hospitalization/surgery) and disease improvement based on the physicians’ global assessment were investigated at baseline, and at 2 and 6 months after eradication or observation. Results A total of 429 IBD (378 ulcerative colitis, 51 Crohn’s disease) patients, comprising 144 patients in the eradication group and 285 patients in the non-eradication group, were enrolled at 25 institutions. IBD exacerbation was comparable between groups (eradication group: 8.3% at 2 months [odds ratio, 1.76; 95% confidence interval, 0.78–3.92; P=0.170], 11.8% at 6 months [odds ratio, 1.60; 95% confidence interval, 0.81–3.11; P=0.172]). Based on the physicians’ global assessment at 2 months, none of the patients in the eradication group improved, whereas 3.2% of the patients in the non-eradication group improved (P=0.019). Multivariate analysis revealed that active disease at baseline, but not H. pylori eradication, was an independent factor for IBD exacerbation during 2 months’ observation period. The overall eradication rate was 84.0%–comparable to previous reports in non-IBD patients. Conclusions H. pylori eradication therapy does not alter the short-term disease activity of IBD.
Collapse
Affiliation(s)
- Shinichiro Shinzaki
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Toshimitsu Fujii
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shigeki Bamba
- Department of Medicine, Shiga University of Medical Science, Otsu, Japan
| | - Maiko Ogawa
- Department of Gastroenterology and Hepatology, The Jikei University School of Medicine Katsushika Medical Center, Tokyo, Japan
| | - Taku Kobayashi
- Center for Advanced IBD Research and Treatment, Kitasato University Kitasato Institute Hospital, Tokyo, Japan
| | - Masahide Oshita
- Department of Internal Medicine, Osaka Police Hospital, Osaka, Japan
| | - Hiroki Tanaka
- IBD Center, Sapporo Kosei General Hospital, Sapporo, Japan
| | - Keiji Ozeki
- Departments of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Sakuma Takahashi
- Department of Gastroenterology, Kagawa Prefectural Central Hospital, Takamatsu, Japan
| | - Hiroki Kitamoto
- Department of Gastroenterology and Hepatology, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Kazuhito Kani
- Department of Gastroenterology and Hepatology, Saitama Medical Center, Saitama Medical University, Kawagoe, Japan
| | - Sohachi Nanjo
- Department of Gastroenterology and Hematology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Takeshi Sugaya
- Department of Gastroenterology, Dokkyo Medical University, Mibu, Japan
| | - Yuko Sakakibara
- Department of Gastroenterology, National Hospital Organization Osaka National Hospital, Osaka, Japan
| | - Toshihiro Inokuchi
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Kazuki Kakimoto
- Second Department of Internal Medicine, Osaka Medical College, Takatsuki, Japan
| | - Akihiro Yamada
- Department of Internal Medicine, Toho University Sakura Medical Center, Sakura, Japan
| | - Hisae Yasuhara
- Department of Gastroenterology, Mitoyo General Hospital, Kanonji, Japan
| | - Yoko Yokoyama
- Department of Inflammatory Bowel Disease, Division of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Takuya Yoshino
- Division of Inflammatory Bowel Disease, Digestive Disease Center, Kitano Hospital, Osaka, Japan
| | - Akira Matsui
- Department of Gastroenterology, Toranomon Hospital, Tokyo, Japan
| | - Misaki Nakamura
- Department of Gastroenterology and Hepatology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Taku Tomizawa
- Department of Gastroenterology and Hepatology, Gunma University, Maebashi, Japan
| | - Ryosuke Sakemi
- Department of Gastroenterology, Tobata Kyoritsu Hospital, Kitakyushu, Japan
| | - Noriko Kamata
- Department of Gastroenterology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Toshifumi Hibi
- Center for Advanced IBD Research and Treatment, Kitasato University Kitasato Institute Hospital, Tokyo, Japan
| |
Collapse
|
89
|
Li J, Li Y, Zhou Y, Wang C, Wu B, Wan J. Actinomyces and Alimentary Tract Diseases: A Review of Its Biological Functions and Pathology. BIOMED RESEARCH INTERNATIONAL 2018; 2018:3820215. [PMID: 30225251 PMCID: PMC6129341 DOI: 10.1155/2018/3820215] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 07/02/2018] [Indexed: 02/06/2023]
Abstract
Actinomyces are nonmotile, filamentous, Gram-positive bacteria that cause actinomycosis in immunodeficiency patients. Although the prognosis of actinomycosis is good, the diagnosis of actinomycosis is quite difficult. Recent studies on actinomycosis have shown that Actinomyces play an important role in various biological and clinical processes, such as the formation of dental plaque and the degradation of organics in the gastrointestinal tract. Here, the distribution of Actinomyces in the digestive tract, and different biological effects of actinomycosis, and its clinical association with inflammatory diseases are discussed. Furthermore, an overview of the most commonly used treatment methods and drugs used to treat Actinomyces infected alimentary canal diseases is presented.
Collapse
Affiliation(s)
- Jun Li
- Department of Gastroenterology, General Hospital of PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Ying Li
- Department of Oncology, General Hospital of PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Yu Zhou
- Department of Nanlou Clinical Laboratory, General Hospital of PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Changzheng Wang
- Department of Gastroenterology, General Hospital of PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Benyan Wu
- Department of Gastroenterology, General Hospital of PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Jun Wan
- Department of Gastroenterology, General Hospital of PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
| |
Collapse
|
90
|
Characterization and prebiotic activity in vitro of inulin-type fructan from Codonopsis pilosula roots. Carbohydr Polym 2018; 193:212-220. [DOI: 10.1016/j.carbpol.2018.03.065] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 03/13/2018] [Accepted: 03/19/2018] [Indexed: 11/19/2022]
|
91
|
Fu YP, Feng B, Zhu ZK, Feng X, Chen SF, Li LX, Yin ZQ, Huang C, Chen XF, Zhang BZ, Jia RY, Song X, Lv C, Yue GZ, Ye G, Liang XX, He CL, Yin LZ, Zou YF. The Polysaccharides from Codonopsis pilosula Modulates the Immunity and Intestinal Microbiota of Cyclophosphamide-Treated Immunosuppressed Mice. Molecules 2018; 23:molecules23071801. [PMID: 30037030 PMCID: PMC6100181 DOI: 10.3390/molecules23071801] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 07/11/2018] [Accepted: 07/19/2018] [Indexed: 01/28/2023] Open
Abstract
Based on previous studies about microflora regulation and immunity enhancement activities of polysaccharides from Codonopsis pilosula Nannf. var. modesta (Nannf.) L. T. Shen (CPP), there is little study on intestinal mucosal immunity, which is a possible medium for contacting microflora and immunity. In the present study, the BALB/c mice were divided into five groups (eight mice in each group), including a normal group (Con), a model control group (Model), and model groups that were administered CPP (50, 100, 200 mg/kg/d) orally each day for seven days after intraperitoneal injection of 60 mg/kg BW/d cyclophosphamide (CP) for three days. CPP recovered the spleen index and restored the levels of IFN-γ, IL-2, IL-10, as well as serum IgG. In addition, it elevated ileum secretory immunoglobulin A (sIgA), the number of Lactobacillus and acetic acid content in cecum. These results indicated that CPP plays an important role in the protection against immunosuppression, especially mucosa immune damage, and the inhibition of pathogenic bacteria colonization, which could be considered a potential natural source of immunoregulator.
Collapse
Affiliation(s)
- Yu-Ping Fu
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China.
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China.
| | - Bin Feng
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China.
| | - Zhong-Kai Zhu
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China.
| | - Xin Feng
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China.
| | - Shu-Fan Chen
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China.
| | - Li-Xia Li
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China.
| | - Zhong-Qiong Yin
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China.
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China.
| | - Chao Huang
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China.
| | - Xing-Fu Chen
- Key Laboratory of Crop Ecophysiology and Farming System in Southwest China, Ministry of Agriculture, College of Agronomy, Sichuan Agricultural University, Chengdu 611130, China.
| | - Bing-Zhao Zhang
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Science, Shenzhen 518055, China.
| | - Ren-Yong Jia
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China.
| | - Xu Song
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China.
| | - Cheng Lv
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China.
| | - Gui-Zhou Yue
- Department of Applied Chemistry, College of Science, Sichuan Agricultural University, Chengdu 611130, China.
| | - Gang Ye
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China.
| | - Xiao-Xia Liang
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China.
| | - Chang-Liang He
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China.
| | - Li-Zi Yin
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China.
| | - Yuan-Feng Zou
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China.
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China.
| |
Collapse
|
92
|
Li J, Chen D, Yu B, He J, Zheng P, Mao X, Yu J, Luo J, Tian G, Huang Z, Luo Y. Fungi in Gastrointestinal Tracts of Human and Mice: from Community to Functions. MICROBIAL ECOLOGY 2018; 75:821-829. [PMID: 29110065 DOI: 10.1007/s00248-017-1105-9] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 10/30/2017] [Indexed: 05/10/2023]
Abstract
Fungi are often ignored in studies on gut microbes because of their low level of presence (making up only 0.1% of the total microorganisms) in the gastrointestinal tract (GIT) of monogastric animals. Recent studies using novel technologies such as next generation sequencing have expanded our understanding on the importance of intestinal fungi in humans and animals. Here, we provide a comprehensive review on the fungal community, the so-called mycobiome, and their functions from recent studies in humans and mice. In the GIT of humans, fungi belonging to the phyla Ascomycota, Basidiomycota and Chytridiomycota are predominant. The murine intestines harbor a more diverse assemblage of fungi. Diet is one of the major factors influencing colonization of fungi in the GIT. Presence of the genus Candida is positively associated with dietary carbohydrates, but are negatively correlated with dietary amino acids, proteins, and fatty acids. However, the relationship between diet and the fungal community (and functions), as well as the underlying mechanisms remains unclear. Dysbiosis of intestinal fungi can cause invasive infections and inflammatory bowel diseases (IBD). However, it is not clear whether dysbiosis of the mycobiome is a cause, or a result of IBD. Compared to non-inflamed intestinal mucosa, the abundance and diversity of fungi is significantly increased in the inflamed mucosa. The commonly observed commensal fungal species Candida albicans might contribute to occurrence and development of IBD. Limited studies show that Candida albicans might interact with immune cells of the host intestines through the pathways associated with Dectin-1, Toll-like receptor 2 (TLR2), and TLR4. This review is expected to provide new thoughts for future studies on intestinal fungi and for new therapies to fungal infections in the GIT of human and animals.
Collapse
Affiliation(s)
- Jiayan Li
- Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Daiwen Chen
- Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Bing Yu
- Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Jun He
- Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Ping Zheng
- Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Xiangbing Mao
- Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Jie Yu
- Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Junqiu Luo
- Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Gang Tian
- Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Zhiqing Huang
- Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Yuheng Luo
- Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China.
| |
Collapse
|
93
|
Solano-Aguilar GI, Lakshman S, Jang S, Beshah E, Xie Y, Sikaroodi M, Gupta R, Vinyard B, Molokin A, Urban JF, Gillevet P, Davis CD. The Effect of Feeding Cocoa Powder and Lactobacillus rhamnosus on the Composition and Function of Pig Intestinal Microbiome. Curr Dev Nutr 2018; 2:nzy011. [PMID: 30019034 PMCID: PMC6041806 DOI: 10.1093/cdn/nzy011] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 08/15/2017] [Accepted: 01/30/2018] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Dietary habits have been linked with variability of gut microbiota composition and disease risk. OBJECTIVE The aim of this study was to evaluate the effect of feeding a cocoa powder with or without a probiotic on the composition and function of the fecal microbiome of pigs. METHODS Four groups of 8 pigs each were fed a standard growth diet supplemented with cocoa powder, Lactobacillus rhamnosus (LGG), cocoa powder + LGG, or an equal amount of fiber similar to that found in cocoa powder (control group). Fecal samples were collected prior to and 4 wk after initiation of the dietary intervention. Microbiota composition was determined after amplification of the first 2 variable regions of the 16S ribosomal DNA (rDNA). Predictions of metagenomic function were calculated using 16S rDNA sequence data through Phylogenetic Investigation of Communities by Reconstruction of Unobserved States (PICRUSt). RESULTS After 4 wk of treatment, bacterial abundance analysis demonstrated a prebiotic effect of cocoa powder on endogenous Bifidobacteriaceae and Lactobacillaceae and increased abundance of saccharolytic butyrate-producing bacteria like Roseburia. An increased bacterial evenness, Shannon diversity index, and diverse metabolic profile were detected in microbiomes of pigs fed the cocoa powder + LGG (P < 0.05) but not in pigs in the other 3 groups. CONCLUSION The data generated from this work demonstrated that 4-wk dietary treatment with cocoa powder alone or in combination with LGG probiotic had an impact on the composition and function of the fecal microbiota of healthy pigs.
Collapse
Affiliation(s)
- Gloria I Solano-Aguilar
- Diet, Genomics and Immunology Laboratory, Beltsville Human Nutrition Research Center, Agricultural Research Service, Northeast Area, US Department of Agriculture, Beltsville, MD
| | - Sukla Lakshman
- Diet, Genomics and Immunology Laboratory, Beltsville Human Nutrition Research Center, Agricultural Research Service, Northeast Area, US Department of Agriculture, Beltsville, MD
| | - Saebyeol Jang
- Diet, Genomics and Immunology Laboratory, Beltsville Human Nutrition Research Center, Agricultural Research Service, Northeast Area, US Department of Agriculture, Beltsville, MD
| | - Ethiopia Beshah
- Diet, Genomics and Immunology Laboratory, Beltsville Human Nutrition Research Center, Agricultural Research Service, Northeast Area, US Department of Agriculture, Beltsville, MD
| | - Yue Xie
- Department of Parasitology, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | | | - Richi Gupta
- Microbiome Analysis Center, George Mason University, Manassas, VA
| | - Bryan Vinyard
- Biometrical Consulting Services, Agricultural Research Service, Northeast Area, US Department of Agriculture, Beltsville, MD
| | - Aleksey Molokin
- Diet, Genomics and Immunology Laboratory, Beltsville Human Nutrition Research Center, Agricultural Research Service, Northeast Area, US Department of Agriculture, Beltsville, MD
| | - Joseph F Urban
- Diet, Genomics and Immunology Laboratory, Beltsville Human Nutrition Research Center, Agricultural Research Service, Northeast Area, US Department of Agriculture, Beltsville, MD
| | - Patrick Gillevet
- Microbiome Analysis Center, George Mason University, Manassas, VA
| | - Cindy D Davis
- Office of Dietary Supplements, National Institutes of Health, Bethesda, MD
| |
Collapse
|
94
|
Abstract
Osteoporosis, characterized by low bone mass and micro-architectural deterioration of bone tissue with increased risk of fracture, can be categorized into two forms: primary and secondary, depending on whether it occurs as part of the natural aging process (estrogen deficiency) or as part of disease pathology. In both forms bone loss is due to an imbalance in the bone remodeling process, with resorption/formation skewed more toward bone loss. Recent studies and emerging evidence consistently demonstrate the potential of the intestinal microbiota to modulate bone health. This review discusses the process of bone remodeling and the pathology of osteoporosis and introduces the intestinal microbiota and its potential to influence bone health. In particular, we highlight recent murine studies that examine how probiotic supplementation can both increase bone density in healthy individuals and protect against primary (estrogen deficiency) as well as secondary osteoporosis. Potential mechanisms are described to account for how probiotic treatments could be exerting their beneficial effect on bone health.
Collapse
|
95
|
Salami AT, Odukanmi OA, Oshode OO, Olaleye SB. Modulatory activities of Chrysophyllum albidum and its fractions on microflora and colonic pump activities during inflammatory phase of colitis healing in experimental mice. FOOD BIOSCI 2018. [DOI: 10.1016/j.fbio.2017.12.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
96
|
McCabe LR, Parameswaran N. Advances in Probiotic Regulation of Bone and Mineral Metabolism. Calcif Tissue Int 2018; 102:480-488. [PMID: 29453726 PMCID: PMC5849527 DOI: 10.1007/s00223-018-0403-7] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 02/08/2018] [Indexed: 12/19/2022]
Abstract
Probiotics have been consumed by humans for thousands of years because they are beneficial for long-term storage of foods and promote the health of their host. Ingested probiotics reside in the gastrointestinal tract where they have many effects including modifying the microbiota composition, intestinal barrier function, and the immune system which result in systemic benefits to the host, including bone health. Probiotics benefit bone growth, density, and structure under conditions of dysbiosis, intestinal permeability, and inflammation (recognized mediators of bone loss and osteoporosis). It is likely that multiple mechanisms are involved in mediating probiotic signals from the gut to the bone. Studies indicate a role for the microbiota (composition and activity), intestinal barrier function, and immune cells in the signaling process. These mechanisms are not mutually exclusive, but rather, may synergize to provide benefits to the skeletal system of the host and serve as a starting point for investigation. Given that probiotics hold great promise for supporting bone health and are generally regarded as safe, future studies identifying mechanisms are warranted.
Collapse
Affiliation(s)
- Laura R McCabe
- Department of Physiology, Michigan State University, East Lansing, MI, USA.
- Department of Radiology, Michigan State University, East Lansing, MI, USA.
- Biomedical Imaging Research Center, Michigan State University, East Lansing, MI, USA.
| | | |
Collapse
|
97
|
Effects of Melatonin on Intestinal Microbiota and Oxidative Stress in Colitis Mice. BIOMED RESEARCH INTERNATIONAL 2018; 2018:2607679. [PMID: 29546052 PMCID: PMC5818891 DOI: 10.1155/2018/2607679] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Revised: 12/29/2017] [Accepted: 01/04/2018] [Indexed: 12/15/2022]
Abstract
This study investigated the antioxidant capacity and intestinal bacteria community in a mouse model of DSS-induced colitis. Twenty mice were randomly assigned to two treatments: mice with colitis induced by 5% DSS (DSS group) and mice with colitis induced by 5% DSS that also received melatonin treatment (MEL group). The DSS group showed significantly less antioxidant capability than the MEL group, but the two groups did not differ significantly in terms of diversity index (Shannon and Simpson), bacterial culture abundance (Chao1 and ACE), and coverage (Good's coverage estimator). Bacteroidetes were the most abundant phylum in the DSS group (58.93%), followed by Firmicutes with 31.46% and Proteobacteria with 7.97%. In contrast, Firmicutes were the most abundant in the MEL group (49.48%), followed by Bacteroidetes with 41.63% and Proteobacteria with 7.50%. The results support the use of melatonin for prevention of intestinal bowel disease due to its modulatory effect on antioxidant capability and microbiota in mice with colitis. Melatonin was demonstrated to improve the oxidative stress resistance of mice with colitis and regulate the intestinal microbial flora, thus improving intestinal health.
Collapse
|
98
|
Alteber Z, Sharbi-Yunger A, Pevsner-Fischer M, Blat D, Roitman L, Tzehoval E, Elinav E, Eisenbach L. The anti-inflammatory IFITM genes ameliorate colitis and partially protect from tumorigenesis by changing immunity and microbiota. Immunol Cell Biol 2018; 96:284-297. [PMID: 29356071 DOI: 10.1111/imcb.12000] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 12/04/2017] [Accepted: 12/06/2017] [Indexed: 12/19/2022]
Abstract
Inflammation plays pivotal roles in different stages of tumor development. Screening for predisposing genetic abnormalities and understanding the roles these genes play in the crosstalk between immune and cancer cells will provide new targets for cancer therapy and prevention. The interferon inducible transmembrane (IFITM) genes are involved in pathogenesis of the gastro-intestinal tract. We aimed at delineating the role of IFITM3 in colonic epithelial homeostasis, inflammation and colitis-associated tumorigenesis using IFITM3-deficient mice. Chemical induction of colitis in IFITM3-deficient mice results in significantly increased clinical signs of inflammation and induction of invasive tumorigenesis. Bone marrow transplantation showed that cells of the hematopoietic system are responsible for colitis deterioration. In these mice, impaired cytokine expression skewed inflammatory response toward pathogenic Th17 with reduced expression of the anti-inflammatory cytokine IL10 during the recovery phase. Intriguingly, mice lacking the entire IFITM locus developed spontaneous chronic colitis from the age of 14 weeks. Sequencing the 16S rRNA of naïve mice lacking IFITM3 gene, or the entire locus containing five IFITM genes, revealed these mice had significant bacterial differences from their wild-type littermates. Our novel results provide strong evidence for the essential role of IFITM genes in ameliorating colitis and colitis-associated tumorigenesis.
Collapse
Affiliation(s)
- Zoya Alteber
- Department of Immunology, The Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Adi Sharbi-Yunger
- Department of Immunology, The Weizmann Institute of Science, Rehovot, 76100, Israel
| | | | - Dan Blat
- Department of Immunology, The Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Lior Roitman
- Department of Immunology, The Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Esther Tzehoval
- Department of Immunology, The Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Eran Elinav
- Department of Immunology, The Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Lea Eisenbach
- Department of Immunology, The Weizmann Institute of Science, Rehovot, 76100, Israel
| |
Collapse
|
99
|
Shi Y, Zhou J, Jiang B, Miao M. Resveratrol and inflammatory bowel disease. Ann N Y Acad Sci 2017; 1403:38-47. [PMID: 28945937 DOI: 10.1111/nyas.13426] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 05/31/2017] [Accepted: 06/07/2017] [Indexed: 12/19/2022]
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory disease of the gastrointestinal tract, comprising ulcerative colitis (UC) and Crohn's disease (CD). Progression of IBD leads to long-term impairment of intestinal structure and function. The pathogenesis of IBD is complex, involving environmental, immunological, genetic, microbial, and psychological factors. The conventional therapies and many existing biopharmaceuticals for IBD have limited efficacy or adverse effects. As a promising safe and effective therapy for IBD, resveratrol has been studied widely, as it has shown anti-inflammatory and antioxidant activity. Resveratrol's mechanism of action involves multiple immune responses and signaling pathways; it is absorbed quickly and metabolized into various derivatives. However, the poor water solubility and low bioavailability of resveratrol limit its clinical applications. Further research should attempt to improve the stability and oral bioavailability of resveratrol by modification and various delivery systems.
Collapse
Affiliation(s)
- Yaning Shi
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, P. R. China.,State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Jie Zhou
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, P. R. China
| | - Bo Jiang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Ming Miao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| |
Collapse
|
100
|
Xing T, Camacho Salazar R, Chen YH. Animal models for studying epithelial barriers in neonatal necrotizing enterocolitis, inflammatory bowel disease and colorectal cancer. Tissue Barriers 2017; 5:e1356901. [PMID: 28795875 DOI: 10.1080/21688370.2017.1356901] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The intestinal epithelial cells line the luminal surface of the entire gastrointestinal tract which is crucial for the absorption of nutrients and prevention of pathogens entering from the external environment. The epithelial barrier plays an important role in organ development, disease pathogenesis, and aging. The major component of an epithelial barrier is the single columnar epithelium and tight junctions. Tight junctions are located at the most apical region of the junctional complex and contain many integral membrane proteins, such as occludin, the claudin family, and junctional adhesion molecules (JAMs). The disruption of intestinal epithelial barriers may lead to several pathophysiological conditions causing malabsorption of nutrition and chronic inflammation. In this review, we provide an update on the alterations of epithelial barriers associated with gut diseases using experimental animal models; we appraise the role of tight junctions in neonatal necrotizing enterocolitis (NEC), inflammatory bowel disease (IBD), and colorectal cancer; we also compare some common features as well as differences and similarities in the pathophysiology of intestinal inflammation in neonatal (NEC) and adult (IBD) gut.
Collapse
Affiliation(s)
- Tiaosi Xing
- a Department of Anatomy and Cell Biology , Brody School of Medicine, East Carolina University , Greenville , NC , USA
| | - Rolando Camacho Salazar
- b Department of Pediatrics , Brody School of Medicine, East Carolina University , Greenville , NC , USA
| | - Yan-Hua Chen
- a Department of Anatomy and Cell Biology , Brody School of Medicine, East Carolina University , Greenville , NC , USA.,b Department of Pediatrics , Brody School of Medicine, East Carolina University , Greenville , NC , USA
| |
Collapse
|