101
|
Early intervention with faecal microbiota transplantation: an effective means to improve growth performance and the intestinal development of suckling piglets. Animal 2018; 13:533-541. [PMID: 29983136 DOI: 10.1017/s1751731118001611] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Recent studies indicate that early postnatal period is a critical window for gut microbiota manipulation to optimise the immunity and body growth. This study investigated the effects of maternal faecal microbiota orally administered to neonatal piglets after birth on growth performance, selected microbial populations, intestinal permeability and the development of intestinal mucosal immune system. In total, 12 litters of crossbred newborn piglets were selected in this study. Litter size was standardised to 10 piglets. On day 1, 10 piglets in each litter were randomly allotted to the faecal microbiota transplantation (FMT) and control groups. Piglets in the FMT group were orally administrated with 2ml faecal suspension of their nursing sow per day from the age of 1 to 3 days; piglets in the control group were treated with the same dose of a placebo (0.1M potassium phosphate buffer containing 10% glycerol (vol/vol)) inoculant. The experiment lasted 21 days. On days 7, 14 and 21, plasma and faecal samples were collected for the analysis of growth-related hormones and cytokines in plasma and lipocalin-2, secretory immunoglobulin A (sIgA), selected microbiota and short-chain fatty acids (SCFAs) in faeces. Faecal microbiota transplantation increased the average daily gain of piglets during week 3 and the whole experiment period. Compared with the control group, the FMT group had increased concentrations of plasma growth hormone and IGF-1 on days 14 and 21. Faecal microbiota transplantation also reduced the incidence of diarrhoea during weeks 1 and 3 and plasma concentrations of zonulin, endotoxin and diamine oxidase activities in piglets on days 7 and 14. The populations of Lactobacillus spp. and Faecalibacterium prausnitzii and the concentrations of faecal and plasma acetate, butyrate and total SCFAs in FMT group were higher than those in the control group on day 21. Moreover, the FMT piglets have higher concentrations of plasma transforming growth factor-β and immunoglobulin G, and faecal sIgA than the control piglets on day 21. These findings indicate that early intervention with maternal faecal microbiota improves growth performance, decreases intestinal permeability, stimulates sIgA secretion, and modulates gut microbiota composition and metabolism in suckling piglets.
Collapse
|
102
|
Guo J, Zhao Y, Jiang X, Li R, Xie H, Ge L, Xie B, Yang X, Zhang L. Exposure to Formaldehyde Perturbs the Mouse Gut Microbiome. Genes (Basel) 2018; 9:E192. [PMID: 29614050 PMCID: PMC5924534 DOI: 10.3390/genes9040192] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 03/20/2018] [Accepted: 03/28/2018] [Indexed: 12/11/2022] Open
Abstract
Exposure to Formaldehyde (FA) results in many pathophysiological symptoms, however the underlying mechanisms are not well understood. Given the complicated modulatory role of intestinal microbiota on human health, we hypothesized that interactions between FA and the gut microbiome may account for FA's toxicity. Balb/c mice were allocated randomly to three groups: a control group, a methanol group (0.1 and 0.3 ng/mL MeOH subgroups), and an FA group (1 and 3 ng/mL FA subgroups). Groups of either three or six mice were used for the control or experiment. We applied high-throughput sequencing of 16S ribosomal RNA (rRNA) gene approaches and investigated possible alterations in the composition of mouse gut microbiota induced by FA. Changes in bacterial genera induced by FA exposure were identified. By analyzing KEGG metabolic pathways predicted by PICRUSt software, we also explored the potential metabolic changes, such as alpha-Linolenic acid metabolism and pathways in cancer, associated with FA exposure in mice. To the best of our knowledge, this preliminary study is the first to identify changes in the mouse gut microbiome after FA exposure, and to analyze the relevant potential metabolisms. The limitation of this study: this study is relatively small and needs to be further confirmed through a larger study.
Collapse
Affiliation(s)
- Junhui Guo
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, China.
- Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley, CA 94720, USA.
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan 430070, China.
| | - Yun Zhao
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, China.
| | - Xingpeng Jiang
- School of Computer, Central China Normal University, Wuhan 430079, China.
| | - Rui Li
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, China.
| | - Hao Xie
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan 430070, China.
| | - Leixin Ge
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, China.
| | - Bo Xie
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, China.
| | - Xu Yang
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, China.
| | - Luoping Zhang
- Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley, CA 94720, USA.
| |
Collapse
|
103
|
Mohammed A, Jacobs J, Murugesan G, Cheng H. Effect of dietary synbiotic supplement on behavioral patterns and growth performance of broiler chickens reared under heat stress. Poult Sci 2018; 97:1101-1108. [DOI: 10.3382/ps/pex421] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Accepted: 12/05/2017] [Indexed: 12/27/2022] Open
|
104
|
Yoshimura T, McLean MH, Dzutsev AK, Yao X, Chen K, Huang J, Gong W, Zhou J, Xiang Y, H Badger J, O'hUigin C, Thovarai V, Tessarollo L, Durum SK, Trinchieri G, Bian XW, Wang JM. The Antimicrobial Peptide CRAMP Is Essential for Colon Homeostasis by Maintaining Microbiota Balance. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 200:2174-2185. [PMID: 29440355 PMCID: PMC5931736 DOI: 10.4049/jimmunol.1602073] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 12/21/2017] [Indexed: 01/05/2023]
Abstract
Commensal bacteria are critical for physiological functions in the gut, and dysbiosis in the gut may cause diseases. In this article, we report that mice deficient in cathelin-related antimicrobial peptide (CRAMP) were defective in the development of colon mucosa and highly sensitive to dextran sulfate sodium (DSS)-elicited colitis, as well as azoxymethane-mediated carcinogenesis. Pretreatment of CRAMP-/- mice with antibiotics markedly reduced the severity of DSS-induced colitis, suggesting CRAMP as a limiting factor on dysbiosis in the colon. This was supported by observations that wild-type (WT) mice cohoused with CRAMP-/- mice became highly sensitive to DSS-induced colitis, and the composition of fecal microbiota was skewed by CRAMP deficiency. In particular, several bacterial species that are typically found in oral microbiota, such as Mogibacterium neglectum, Desulfovibrio piger, and Desulfomicrobium orale, were increased in feces of CRAMP-/- mice and were transferred to WT mice during cohousing. When littermates of CRAMP+/- parents were examined, the composition of the fecal microbiota of WT pups and heterozygous parents was similar. In contrast, although the difference in fecal microbiota between CRAMP-/- and WT pups was small early on after weaning and single mouse housing, there was an increasing divergence with prolonged single housing. These results indicate that CRAMP is critical in maintaining colon microbiota balance and supports mucosal homeostasis, anti-inflammatory responses, and protection from carcinogenesis.
Collapse
Affiliation(s)
- Teizo Yoshimura
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702;
- Department of Pathology and Experimental Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan
| | - Mairi H McLean
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, United Kingdom
| | - Amiran K Dzutsev
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702
| | - Xiaohong Yao
- Institute of Pathology and Southwest Cancer Center, Third Military Medical University, Chongqing 400038, China
| | - Keqiang Chen
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702
| | - Jiaqiang Huang
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702
- College of Life Sciences and Bioengineering, School of Science, Beijing Jiaotong University, Beijing 100044, China
| | - Wanghua Gong
- Basic Science Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702; and
| | - Jiamin Zhou
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702
| | - Yi Xiang
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702
| | - Jonathan H Badger
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702
| | - Colm O'hUigin
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702
| | - Vishal Thovarai
- Basic Science Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702; and
| | - Lino Tessarollo
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702
| | - Scott K Durum
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702
| | - Giorgio Trinchieri
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702
| | - Xiu-Wu Bian
- Institute of Pathology and Southwest Cancer Center, Third Military Medical University, Chongqing 400038, China
| | - Ji Ming Wang
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702;
| |
Collapse
|
105
|
Farhat-Khemakhem A, Blibech M, Boukhris I, Makni M, Chouayekh H. Assessment of the potential of the multi-enzyme producer Bacillus amyloliquefaciens US573 as alternative feed additive. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2018; 98:1208-1215. [PMID: 28741666 DOI: 10.1002/jsfa.8574] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 07/11/2017] [Accepted: 07/20/2017] [Indexed: 06/07/2023]
Abstract
BACKGROUND Recently, probiotics have increasingly been used as feed additives in poultry diets as an alternative to antibiotic growth promoters fostering resistance development. RESULTS This study was aimed at assessing the potential of Bacillus amyloliquefaciens US573 as a direct-fed microbial. The US573 strain was found to be free of harmful enzymatic activities and sensitive to antibiotics. In addition, it showed a good acid and bovine bile tolerance, high adhesion efficacy to chicken enterocytes, and an ability to form biofilms, which may favor its survival and persistence in the animal gastrointestinal tract. Moreover, besides the previously described extremely salt-tolerant and highly thermostable phytase, the US573 strain secretes xylanase, β-glucanase and amylase activities useful in neutralizing antinutritional factors and maximizing the absorption of nutrients. The secretion of such enzymes may be responsible for the good performance of the US573 isolate in the digestibility of wheat in vitro. Indeed, using the vegetative cells, a yield of wheat dry matter digestibility of approximately 48% was achieved, which is slightly lower than the commercial feed additive Rovabio used as a reference (56.73% digestibility). CONCLUSION The obtained results illustrate the potential of US573 strain as a promising direct-fed microbial candidate for application in the poultry industry. © 2017 Society of Chemical Industry.
Collapse
Affiliation(s)
- Ameny Farhat-Khemakhem
- Laboratoire de Microorganismes et de Biomolécules, Centre de Biotechnologie de Sfax, Université de Sfax, BP, 1177, 3018, Sfax, Tunisia
| | - Monia Blibech
- Laboratoire de Microorganismes et de Biomolécules, Centre de Biotechnologie de Sfax, Université de Sfax, BP, 1177, 3018, Sfax, Tunisia
| | - Ines Boukhris
- Laboratoire de Microorganismes et de Biomolécules, Centre de Biotechnologie de Sfax, Université de Sfax, BP, 1177, 3018, Sfax, Tunisia
| | - Mohamed Makni
- Laboratoire de Physiologie Animale, Faculté des Sciences de Sfax, BP, 1171, 3000, Sfax, Tunisia
| | - Hichem Chouayekh
- Laboratoire de Microorganismes et de Biomolécules, Centre de Biotechnologie de Sfax, Université de Sfax, BP, 1177, 3018, Sfax, Tunisia
| |
Collapse
|
106
|
|
107
|
Voigt RM, Forsyth CB, Shaikh M, Zhang L, Raeisi S, Aloman C, Preite NZ, Donohue TM, Fogg L, Keshavarzian A. Diurnal variations in intestinal barrier integrity and liver pathology in mice: implications for alcohol binge. Am J Physiol Gastrointest Liver Physiol 2018; 314:G131-G141. [PMID: 29074484 PMCID: PMC5866370 DOI: 10.1152/ajpgi.00103.2017] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 10/12/2017] [Accepted: 10/13/2017] [Indexed: 02/08/2023]
Abstract
Recent studies suggest that circadian rhythms regulate intestinal barrier integrity, but it is not clear whether there are daily variations in barrier integrity. This study investigated daily variations in intestinal barrier integrity, including whether there are differences in alcohol-induced intestinal barrier dysfunction after an alcohol binge at different times of day and whether this is associated with concurrent liver injury. C57BL6/J male mice were fed a standard chow diet, an alcohol-containing liquid diet, or an alcohol control diet for 4 wk. During week 5 (i.e., on days 43-45), mice received three once-daily gavages of alcohol (6 g/kg) or the control (phosphate-buffered saline) at the same time each day. Immediately after the binge on the second day, intestinal permeability was assessed. Four hours after the third and final binge, mice were euthanized and tissue samples collected. The results demonstrated diet-specific and outcome-specific effects of time, alcohol, and/or time by alcohol interaction. Specifically, the alcohol binge robustly influenced markers of intestinal barrier integrity, and liver markers were robustly influenced by time of day. Only intestinal permeability (i.e., sucralose) demonstrated a significant effect of time and also showed a binge by time interaction, suggesting that the time of the alcohol binge influences colonic permeability. NEW & NOTEWORTHY This study investigated daily variations in intestinal barrier integrity, including whether there are differences in alcohol-induced intestinal barrier dysfunction after an alcohol binge at different times of day and whether this is associated with concurrent liver injury. We conclude that 1) alcohol binge significantly impacted markers of intestinal permeability, 2) time of day significantly affected liver outcomes, and 3) the time of day influenced colonic permeability.
Collapse
Affiliation(s)
- Robin M Voigt
- Section of Gastroenterology, Division of Digestive Diseases and Nutrition, Department of Internal Medicine, Rush University Medical Center , Chicago, Illinois
| | - Christopher B Forsyth
- Section of Gastroenterology, Division of Digestive Diseases and Nutrition, Department of Internal Medicine, Rush University Medical Center , Chicago, Illinois
- Department of Biochemistry, Rush University , Chicago, Illinois
| | - Maliha Shaikh
- Section of Gastroenterology, Division of Digestive Diseases and Nutrition, Department of Internal Medicine, Rush University Medical Center , Chicago, Illinois
| | - Lijuan Zhang
- Section of Gastroenterology, Division of Digestive Diseases and Nutrition, Department of Internal Medicine, Rush University Medical Center , Chicago, Illinois
| | - Shohreh Raeisi
- Section of Gastroenterology, Division of Digestive Diseases and Nutrition, Department of Internal Medicine, Rush University Medical Center , Chicago, Illinois
| | - Costica Aloman
- Section of Gastroenterology, Division of Digestive Diseases and Nutrition, Department of Internal Medicine, Rush University Medical Center , Chicago, Illinois
| | - Nailliw Z Preite
- Section of Gastroenterology, Division of Digestive Diseases and Nutrition, Department of Internal Medicine, Rush University Medical Center , Chicago, Illinois
| | - Terrence M Donohue
- Departments of Internal Medicine and Biochemistry/Molecular Biology, University of Nebraska Medical Center , Omaha, Nebraska
| | - Louis Fogg
- Community, Systems and Mental Health Nursing, Rush University Medical Center , Chicago, Illinois
| | - Ali Keshavarzian
- Section of Gastroenterology, Division of Digestive Diseases and Nutrition, Department of Internal Medicine, Rush University Medical Center , Chicago, Illinois
- Department of Pharmacology, Rush University , Chicago, Illinois
- Department of Molecular Biophysics and Physiology, Rush University , Chicago, Illinois
- F. C. Donders Chair, Faculty of Science, University of Utrecht , Utrecht , The Netherlands
| |
Collapse
|
108
|
Fernandes SM, Pires AR, Matoso P, Ferreira C, Nunes-Cabaço H, Correia L, Valadas E, Poças J, Pacheco P, Veiga-Fernandes H, Foxall RB, Sousa AE. HIV-2 infection is associated with preserved GALT homeostasis and epithelial integrity despite ongoing mucosal viral replication. Mucosal Immunol 2018; 11:236-248. [PMID: 28513595 DOI: 10.1038/mi.2017.44] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 04/10/2017] [Indexed: 02/04/2023]
Abstract
The mechanisms that enable preservation of gut mucosal integrity during persistent viral replication and inherent inflammation remain unclear. Here, we investigated, for the first time, gut homeostasis in HIV-2 infection, a naturally occurring form of attenuated HIV disease. We found viral replication in both sigmoid and ileum of asymptomatic HIV-2+ patients (range: 240-851 circulating CD4+T-cells per μl) despite their undetectable viremia, accompanied by interferon-γ-producing CD8 T-cell expansion, irrespective of antiretroviral treatment. Nevertheless, there was no CD4 T-cell depletion, and Foxp3+ and IL-17- or IL-22-producing CD4 T-cell numbers were unaffected. Moreover, IL-22-producing innate lymphoid cells and IL-22-induced antimicrobial peptides and mucins were maintained. In agreement, the epithelium histology was preserved, including tight junction protein zonula occludens (ZO-1) levels. Furthermore, in vitro infection of colon epithelia with primary isolates revealed no HIV-2 impact on ZO-1 expression. Notably, sigmoid transcriptional levels of CCL20 and CCL28 were significantly increased, in direct correlation with GM-CSF, indicating a local response able to enhance CD4 T-cell recruitment. In conclusion, maintenance of mucosal integrity in HIV-2 infection was associated with T-cell recruitment responses, potentially counteracting CD4 T-cell depletion due to HIV-2 replication. These data have unique implications for the design of therapies targeting gut homeostasis in HIV-1 infection and other chronic inflammatory settings.
Collapse
Affiliation(s)
- S M Fernandes
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Hospital de Santa Maria, Centro Hospitalar Lisboa Norte-EPE, Lisboa, Portugal
| | - A R Pires
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - P Matoso
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - C Ferreira
- Hospital de Santa Maria, Centro Hospitalar Lisboa Norte-EPE, Lisboa, Portugal
| | - H Nunes-Cabaço
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - L Correia
- Hospital de Santa Maria, Centro Hospitalar Lisboa Norte-EPE, Lisboa, Portugal
| | - E Valadas
- Hospital de Santa Maria, Centro Hospitalar Lisboa Norte-EPE, Lisboa, Portugal
- Clínica Universitária de Doenças Infecciosas, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - J Poças
- Serviço de Infecciologia, Hospital de S. Bernardo, Setúbal, Portugal
| | - P Pacheco
- Serviço de Infecciologia, Hospital Fernando da Fonseca, Amadora, Portugal
| | - H Veiga-Fernandes
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - R B Foxall
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - A E Sousa
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
109
|
Gastrointestinal Symptoms and Elevated Levels of Anti- Saccharomyces cerevisiae Antibodies Are Associated with Higher Disease Activity in Colombian Patients with Spondyloarthritis. Int J Rheumatol 2017; 2017:4029584. [PMID: 29213287 PMCID: PMC5682082 DOI: 10.1155/2017/4029584] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 07/10/2017] [Accepted: 08/02/2017] [Indexed: 12/26/2022] Open
Abstract
Background Spondyloarthritis (SpA) is a group of articular inflammatory rheumatic diseases that their gastrointestinal manifestations are around 10% of their extra-articular symptoms, supporting that the inflammatory response of the intestinal mucosa could be associated with the clinical status. Objectives To investigate the association between gastrointestinal symptoms and autoantibodies and disease activity between SpA patients, healthy subjects (HS), and patients with inflammatory bowel disease (IBD). Methods 102 SpA patients, 29 IBD patients, and 117 HS were included. Autoantibodies as ASCA, ANCA, anti-tTG, anti-DGP, ANA, and IgA were measured. The patients were assessed to evaluate clinical and gastrointestinal symptoms. An association analysis was performed using Chi square test and a logistic regression. Results Significant differences were found for ASCA levels in SpA (28.2%) compared to IBD (14.2%) and HS (6.0%) (p = 0.029), as well as for ANAS in SpA (49.5%) and IBD (37.9%) (p < 0.001) and abdominal pain (p = 0.012) between SpA (54.3%) and IBD (27.5%). Significant associations were found between BASDAI > 4 and gastrointestinal symptoms (p < 0.05) and IgA (p = 0.007). The association for abdominal bloating was maintained (OR: 3.93, CI-95%, 1.14–13.56; p = 0.030). Conclusions Gastrointestinal symptoms, ASCA, ANAS, and IgA levels were associated with high disease activity in SpA compared with IBD and HS.
Collapse
|
110
|
Yadav M, Verma MK, Chauhan NS. A review of metabolic potential of human gut microbiome in human nutrition. Arch Microbiol 2017; 200:203-217. [PMID: 29188341 DOI: 10.1007/s00203-017-1459-x] [Citation(s) in RCA: 144] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 10/30/2017] [Accepted: 11/16/2017] [Indexed: 02/06/2023]
Abstract
The human gut contains a plethora of microbes, providing a platform for metabolic interaction between the host and microbiota. Metabolites produced by the gut microbiota act as a link between gut microbiota and its host. These metabolites act as messengers having the capacity to alter the gut microbiota. Recent advances in the characterization of the gut microbiota and its symbiotic relationship with the host have provided a platform to decode metabolic interactions. The human gut microbiota, a crucial component for dietary metabolism, is shaped by the genetic, epigenetic and dietary factors. The metabolic potential of gut microbiota explains its significance in host health and diseases. The knowledge of interactions between microbiota and host metabolism, as well as modification of microbial ecology, is really beneficial to have effective therapeutic treatments for many diet-related diseases in near future. This review cumulates the information to map the role of human gut microbiota in dietary component metabolism, the role of gut microbes derived metabolites in human health and host-microbe metabolic interactions in health and diseases.
Collapse
Affiliation(s)
- Monika Yadav
- Department of Biochemistry, Maharshi Dayanand University, Rohtak, Haryana, 124001, India
| | - Manoj Kumar Verma
- Department of Biochemistry, Maharshi Dayanand University, Rohtak, Haryana, 124001, India
| | - Nar Singh Chauhan
- Department of Biochemistry, Maharshi Dayanand University, Rohtak, Haryana, 124001, India.
| |
Collapse
|
111
|
Takiishi T, Fenero CIM, Câmara NOS. Intestinal barrier and gut microbiota: Shaping our immune responses throughout life. Tissue Barriers 2017; 5:e1373208. [PMID: 28956703 DOI: 10.1080/21688370.2017.1373208] [Citation(s) in RCA: 604] [Impact Index Per Article: 75.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The gastrointestinal (GI) tract is considered the largest immunological organ in the body having a central role in regulating immune homeostasis. Contrary to earlier belief, the intestinal epithelial barrier is not a static physical barrier but rather strongly interacts with the gut microbiome and cells of the immune system. This intense communication between epithelial cells, immune cells and microbiome will shape specific immune responses to antigens, balancing tolerance and effector immune functions. Recent studies indicate that composition of the gut microbiome affects immune system development and modulates immune mediators, which in turn affect the intestinal barrier. Moreover, dysbiosis may favor intestinal barrier disruption and could be related to increased susceptibility to certain diseases. This review will be focused on the development of the intestinal barrier and its function in host immune defense and how gut microbiome composition throughout life can affect this role.
Collapse
Affiliation(s)
- Tatiana Takiishi
- a Department of Immunology, Institute of Biomedical Sciences , University of São Paulo (USP), São Paulo - SP , Brazil
| | - Camila Ideli Morales Fenero
- a Department of Immunology, Institute of Biomedical Sciences , University of São Paulo (USP), São Paulo - SP , Brazil
| | - Niels Olsen Saraiva Câmara
- a Department of Immunology, Institute of Biomedical Sciences , University of São Paulo (USP), São Paulo - SP , Brazil
| |
Collapse
|
112
|
Abstract
The gastrointestinal mucosa constitutes a critical barrier where millions of microbes and environmental antigens come in close contact with the host immune system. Intestinal barrier defects have been associated with a broad range of diseases and therefore denote a new therapeutic target. Areas covered: This review is based on an extensive literature search in PubMed of how the intestinal barrier contributes to health and as a trigger for disease. It discusses the anatomy of the intestinal barrier and explains the available methods to evaluate its function. Also reviewed is the importance of diet and lifestyle factors on intestinal barrier function, and three prototypes of chronic diseases (inflammatory bowel disease, celiac disease and nonalcoholic fatty liver disease) that have been linked to barrier defects are discussed. Expert commentary: The intestinal barrier has been investigated by various methods, but correlation of results across studies is difficult, representing a major shortcoming in the field. New upcoming techniques and research on the effect of barrier-restoring therapeutics may improve our current understanding of the gut barrier, and provide a step forward towards personalised medicine.
Collapse
|
113
|
Weiss GA, Hennet T. Mechanisms and consequences of intestinal dysbiosis. Cell Mol Life Sci 2017; 74:2959-2977. [PMID: 28352996 PMCID: PMC11107543 DOI: 10.1007/s00018-017-2509-x] [Citation(s) in RCA: 425] [Impact Index Per Article: 53.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 03/08/2017] [Accepted: 03/15/2017] [Indexed: 02/07/2023]
Abstract
The composition of the gut microbiota is in constant flow under the influence of factors such as the diet, ingested drugs, the intestinal mucosa, the immune system, and the microbiota itself. Natural variations in the gut microbiota can deteriorate to a state of dysbiosis when stress conditions rapidly decrease microbial diversity and promote the expansion of specific bacterial taxa. The mechanisms underlying intestinal dysbiosis often remain unclear given that combinations of natural variations and stress factors mediate cascades of destabilizing events. Oxidative stress, bacteriophages induction and the secretion of bacterial toxins can trigger rapid shifts among intestinal microbial groups thereby yielding dysbiosis. A multitude of diseases including inflammatory bowel diseases but also metabolic disorders such as obesity and diabetes type II are associated with intestinal dysbiosis. The characterization of the changes leading to intestinal dysbiosis and the identification of the microbial taxa contributing to pathological effects are essential prerequisites to better understand the impact of the microbiota on health and disease.
Collapse
Affiliation(s)
- G Adrienne Weiss
- Institute of Physiology, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
| | - Thierry Hennet
- Institute of Physiology, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland.
| |
Collapse
|
114
|
Abstract
Microbiota research, in particular that of the gut, has recently gained much attention in medical research owing to technological advances in metagenomics and metabolomics. Despite this, much of the research direction has focused on long-term or chronic effects of microbiota manipulation on health and disease. In this addendum, we reflect on our recent publication that reported findings addressing a rather unconventional hypothesis. Bacterial pneumonia is highly prevalent and is one of the leading contributors to stroke morbidity and mortality worldwide. However, microbiological cultures of samples taken from stroke patient with a suspected case of pneumonia often return with a negative result. Therefore, we proposed that post-stroke infection may be due to the presence of anaerobic bacteria, possibly those originated from the host gut microbiota. Supporting this, we showed that stroke promotes intestinal barrier breakdown and robust microbiota changes, and the subsequent translocation of selective bacterial strain from the host gut microbiota to peripheral tissues (i.e. lung) induces post-stroke infections. Our findings were further supported by various elegant studies published in the past 12 months. Here, we discuss and provide an overview of our key findings, supporting studies, and the implications for future advances in stroke research.
Collapse
Affiliation(s)
- Shu Wen Wen
- Centre for Inflammatory Diseases, Department of Medicine, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia
| | - Connie H. Y. Wong
- Centre for Inflammatory Diseases, Department of Medicine, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia,CONTACT Connie H. Y. Wong, PhD. , Centre for Inflammatory Diseases, Department of Medicine, School of Clinical Sciences, Monash Medical Centre, Monash University, Clayton, VIC 3168 Australia
| |
Collapse
|
115
|
Abstract
This narrative review summarises the benefits, risks and appropriate use of acid-suppressing drugs (ASDs), proton pump inhibitors and histamine-2 receptor antagonists, advocating a rationale balanced and individualised approach aimed to minimise any serious adverse consequences. It focuses on current controversies on the potential of ASDs to contribute to infections-bacterial, parasitic, fungal, protozoan and viral, particularly in the elderly, comprehensively and critically discusses the growing body of observational literature linking ASD use to a variety of enteric, respiratory, skin and systemic infectious diseases and complications (Clostridium difficile diarrhoea, pneumonia, spontaneous bacterial peritonitis, septicaemia and other). The proposed pathogenic mechanisms of ASD-associated infections (related and unrelated to the inhibition of gastric acid secretion, alterations of the gut microbiome and immunity), and drug-drug interactions are also described. Both probiotics use and correcting vitamin D status may have a significant protective effect decreasing the incidence of ASD-associated infections, especially in the elderly. Despite the limitations of the existing data, the importance of individualised therapy and caution in long-term ASD use considering the balance of benefits and potential harms, factors that may predispose to and actions that may prevent/attenuate adverse effects is evident. A six-step practical algorithm for ASD therapy based on the best available evidence is presented.
Collapse
Affiliation(s)
- Leon Fisher
- Frankston Hospital, Peninsula Health, Melbourne, Australia.
| | - Alexander Fisher
- The Canberra Hospital, ACT Health, Canberra, Australia
- Australian National University Medical School, Canberra, Australia
| |
Collapse
|
116
|
Tumor Necrosis Factor α-Dependent Neutrophil Priming Prevents Intestinal Ischemia/Reperfusion-Induced Bacterial Translocation. Dig Dis Sci 2017; 62:1498-1510. [PMID: 28144894 DOI: 10.1007/s10620-017-4468-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 01/20/2017] [Indexed: 02/08/2023]
Abstract
BACKGROUND Intestinal ischemia/reperfusion (I/R) causes barrier impairment and bacterial influx. Protection against I/R injury in sterile organs by hypoxic preconditioning (HPC) had been attributed to erythropoietic and angiogenic responses. Our previous study showed attenuation of intestinal I/R injury by HPC for 21 days in a neutrophil-dependent manner. AIM To investigate the underlying mechanisms of neutrophil priming by HPC, and explore whether adoptive transfer of primed neutrophils is sufficient to ameliorate intestinal I/R injury. METHODS Rats raised in normoxia (NM) and HPC for 3 or 7 days were subjected to sham operation or superior mesenteric artery occlusion for I/R challenge. Neutrophils isolated from rats raised in NM or HPC for 21 days were intravenously injected into naïve controls prior to I/R. RESULTS Similar to the protective effect of HPC-21d, I/R-induced mucosal damage was attenuated by HPC-7d but not by HPC-3d. Naïve rats reconstituted with neutrophils of HPC-21d rats showed increase in intestinal phagocytic infiltration and myeloperoxidase activity, and barrier protection against I/R insult. Elevated free radical production, and higher bactericidal and phagocytic activity were observed in HPC neutrophils compared to NM controls. Moreover, increased serum levels of tumor necrosis factor α (TNFα) and cytokine-induced neutrophil chemoattractant-1 (CINC-1) were seen in HPC rats. Naïve neutrophils incubated with HPC serum or recombinant TNFα, but not CINC-1, exhibited heightened respiratory burst and bactericidal activity. Lastly, neutrophil priming effect was abolished by neutralization of TNFα in HPC serum. CONCLUSIONS TNFα-primed neutrophils by HPC act as effectors cells for enhancing barrier integrity under gut ischemia.
Collapse
|
117
|
Survival of the Fittest: How Bacterial Pathogens Utilize Bile To Enhance Infection. Clin Microbiol Rev 2017; 29:819-36. [PMID: 27464994 DOI: 10.1128/cmr.00031-16] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Bacterial pathogens have coevolved with humans in order to efficiently infect, replicate within, and be transmitted to new hosts to ensure survival and a continual infection cycle. For enteric pathogens, the ability to adapt to numerous host factors under the harsh conditions of the gastrointestinal tract is critical for establishing infection. One such host factor readily encountered by enteric bacteria is bile, an innately antimicrobial detergent-like compound essential for digestion and nutrient absorption. Not only have enteric pathogens evolved to resist the bactericidal conditions of bile, but these bacteria also utilize bile as a signal to enhance virulence regulation for efficient infection. This review provides a comprehensive and up-to-date analysis of bile-related research with enteric pathogens. From common responses to the unique expression of specific virulence factors, each pathogen has overcome significant challenges to establish infection in the gastrointestinal tract. Utilization of bile as a signal to modulate virulence factor expression has led to important insights for our understanding of virulence mechanisms for many pathogens. Further research on enteric pathogens exposed to this in vivo signal will benefit therapeutic and vaccine development and ultimately enhance our success at combating such elite pathogens.
Collapse
|
118
|
Wang LC, Yu Q, Edwards V, Lin B, Qiu J, Turner JR, Stein DC, Song W. Neisseria gonorrhoeae infects the human endocervix by activating non-muscle myosin II-mediated epithelial exfoliation. PLoS Pathog 2017; 13:e1006269. [PMID: 28406994 PMCID: PMC5391109 DOI: 10.1371/journal.ppat.1006269] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 03/02/2017] [Indexed: 12/24/2022] Open
Abstract
Colonization and disruption of the epithelium is a major infection mechanism of mucosal pathogens. The epithelium counteracts infection by exfoliating damaged cells while maintaining the mucosal barrier function. The sexually transmitted bacterium Neisseria gonorrhoeae (GC) infects the female reproductive tract primarily from the endocervix, causing gonorrhea. However, the mechanism by which GC overcome the mucosal barrier remains elusive. Using a new human tissue model, we demonstrate that GC can penetrate into the human endocervix by inducing the exfoliation of columnar epithelial cells. We found that GC colonization causes endocervical epithelial cells to shed. The shedding results from the disassembly of the apical junctions that seal the epithelial barrier. Apical junction disruption and epithelial exfoliation increase GC penetration into the endocervical epithelium without reducing bacterial adherence to and invasion into epithelial cells. Both epithelial exfoliation and junction disruption require the activation and accumulation of non-muscle myosin II (NMII) at the apical surface and GC adherent sites. GC inoculation activates NMII by elevating the levels of the cytoplasmic Ca2+ and NMII regulatory light chain phosphorylation. Piliation of GC promotes, but the expression of a GC opacity-associated protein variant, OpaH that binds to the host surface proteins CEACAMs, inhibits GC-induced NMII activation and reorganization and Ca2+ flux. The inhibitory effects of OpaH lead to reductions in junction disruption, epithelial exfoliation, and GC penetration. Therefore, GC phase variation can modulate infection in the human endocervix by manipulating the activity of NMII and epithelial exfoliation. Neisseria gonorrhoeae (GC) infects human genital epithelium causing gonorrhea, a common sexually transmitted infection. Gonorrhea is a critical public health issue due to increased prevalence of antibiotic-resistant strains. Because humans are the only host for GC, a lack of a human infection model has been a major obstacle to our understanding of GC infection. Here we use a human tissue explant model to examine the mechanism by which GC infect the human endocervix, the primary site for GC infection in women. We show that GC penetrate into the human endocervix by activating the actin motor myosin and epithelial shedding. Myosin activation causes the disruption of the endocervical epithelial barrier by inducing apical junction disassembly and epithelial cell shedding, allowing GC penetration into the human endocervical tissue. GC activate myosin by inducing Ca2+-dependent phosphorylation of myosin light chain. We further show that GC can enhance and reduce the penetration by expressing pili and the opacity-associated protein that promotes and inhibits myosin activation, respectively. Our study is the first demonstration of GC penetration into the human endocervix. Our results provide new insights into the mechanism by which GC manipulate signaling and cytoskeletal apparatus in epithelial cells to achieve penetrating and non-penetrating infection.
Collapse
Affiliation(s)
- Liang-Chun Wang
- Department of Cell Biology & Molecular Genetics, University of Maryland, College Park, Maryland, United States of America
| | - Qian Yu
- Department of Cell Biology & Molecular Genetics, University of Maryland, College Park, Maryland, United States of America
| | - Vonetta Edwards
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Brian Lin
- Department of Cell Biology & Molecular Genetics, University of Maryland, College Park, Maryland, United States of America
| | - Jessica Qiu
- Department of Cell Biology & Molecular Genetics, University of Maryland, College Park, Maryland, United States of America
| | - Jerrold R. Turner
- Departments of Pathology and Medicine (GI), Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Daniel C. Stein
- Department of Cell Biology & Molecular Genetics, University of Maryland, College Park, Maryland, United States of America
| | - Wenxia Song
- Department of Cell Biology & Molecular Genetics, University of Maryland, College Park, Maryland, United States of America
- * E-mail:
| |
Collapse
|
119
|
Xu JH, Liu XZ, Pan W, Zou DJ. Berberine protects against diet-induced obesity through regulating metabolic endotoxemia and gut hormone levels. Mol Med Rep 2017; 15:2765-2787. [PMID: 28447763 PMCID: PMC5428400 DOI: 10.3892/mmr.2017.6321] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 01/16/2017] [Indexed: 12/19/2022] Open
Abstract
Systemic inflammation, which can be induced by metabolic endotoxemia, and corresponding high‑fat diet‑mediated metabolic disorders are associated with gut microbiota. In the present study reverse transcription-polymerase chain reaction, immunofluorescence, pyrosequencing, ELISA and Oil Red O staining were performed to assess whether berberine can protect against diet-induced obesity, through modulating the gut microbiota and consequently improving metabolic endotoxemia and gastrointestinal hormone levels. Alterations in the gut microbiota induced by berberine resulted in a significant reduction in bacterial lipopolysaccharide levels in portal plasma. Levels of inflammatory and oxidative stress markers, as well as the mRNA expression levels of macrophage infiltration markers in visceral adipose tissue, were also reduced by berberine. Inhibition of the inflammatory response was associated with a reduction in intestinal permeability and an increase in the expression of tight junction proteins. In addition, berberine was reported to restore aberrant levels of gut hormones in the portal plasma, such as glucagon‑like peptide‑1 and ‑2, peptide YY, glucose‑dependent insulinotropic polypeptide and pancreatic polypeptide. The present findings indicated that berberine, through modulating gut microbiota, restored the gut barrier, reduced metabolic endotoxemia and systemic inflammation, and improved gut peptide levels in high‑fat diet‑fed rats. The present study suggests that berberine may be an effective therapeutic strategy for the treatment of obesity and insulin resistance.
Collapse
Affiliation(s)
- Jian Hui Xu
- Department of Endocrinology, Changhai Hospital, The Second Military Medical University, Shanghai 200433, P.R. China
| | - Xing Zhen Liu
- The Out‑Patient Department, Hangzhou Sanatorium of Nanjing Military Command Region, Hangzhou, Zhejiang 310007, P.R. China
| | - Wei Pan
- Department of Pathogen Biology, The Second Military Medical University, Shanghai 200433, P.R. China
| | - Da Jin Zou
- Department of Endocrinology, Changhai Hospital, The Second Military Medical University, Shanghai 200433, P.R. China
| |
Collapse
|
120
|
A Novel Role of Spred2 in the Colonic Epithelial Cell Homeostasis and Inflammation. Sci Rep 2016; 6:37531. [PMID: 27869219 PMCID: PMC5116627 DOI: 10.1038/srep37531] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 10/27/2016] [Indexed: 12/13/2022] Open
Abstract
Rapid and adequate mucosal healing is important for a remission of ulcerative colitis (UC) patients. Here, we examined whether Spred2, a member of the Sprouty-related EVH1-domain-containing proteins that inhibit the Ras/Raf/ERK pathway, plays a role in colonic mucosal homeostasis and inflammation by using Spred2 knockout (KO) mice. We first detected increased epithelial cell proliferation and cadherin 1 expression in the colon of naïve Spred2 KO mice compared to wild-type mice. Interestingly, Spred2 KO mice were resistant to dextran sulfate sodium (DSS)-induced acute colitis as indicated by lower levels of body weight loss and disease activity index. Histologically, epithelial cell injury and inflammation were milder in the colonic mucosa of Spred2 KO mice on day 3 and almost undetectable by day 8. Experiments with bone chimeric mice indicated that Spred2-deficiency in non-hematopoietic cells was responsible for the reduced sensitivity to DSS. Finally, Spred2 KO mice developed significantly fewer tumors in response to azoxymethane plus DSS. Taken together, our results demonstrate, for the first time, that Spred2 plays an important role in the regulation of colonic epithelial cell proliferation and inflammation by potentially down-regulating the activation of ERK. Thus, Spred2 may be a new therapeutic target for the treatment of UC.
Collapse
|
121
|
Loganes C, Pin A, Naviglio S, Girardelli M, Bianco AM, Martelossi S, Tommasini A, Piscianz E. Altered pattern of tumor necrosis factor-alpha production in peripheral blood monocytes from Crohn's disease. World J Gastroenterol 2016; 22:9117-9126. [PMID: 27895399 PMCID: PMC5107593 DOI: 10.3748/wjg.v22.i41.9117] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 08/25/2016] [Accepted: 09/14/2016] [Indexed: 02/06/2023] Open
Abstract
AIM To evaluate the inflammatory state in Crohn's disease (CD) patients and correlate it with genetic background and microbial spreading. METHODS By means of flow cytometry, production of tumor necrosis factor-alpha (TNF-α) was measured in peripheral blood monocytes from patients suffering from CD, ulcerative colitis (UC) and in healthy subjects after stimulation of the NOD2 and TLR pathways. CD patients were genotyped for the three most common NOD2 variants (R702W, G908R and L1007Pfs*2) and basal production of TNF-α was correlated to NOD2 genotype. Also, production of TNF-α was correlated to plasmatic levels of LPS Binding Protein (LBP), soluble (s) CD14 and to the activity state of the disease. RESULTS The patients with CD were characterized by a significantly higher monocyte basal expression of TNF-α compared with healthy subjects and UC patients, and after stimulation with Pam3CSK4 (ligand of TLR2/1) and MDP-L18 (ligand of NOD2) this difference was maintained, while other microbial stimuli (LPS, ligand of TLR4 and PolyI:C, ligand of TLR3) induced massive activation in CD monocytes as well as in UC and in healthy control cells. There was no significant difference in the production of TNF-α between patients who carried CD-associated heterozygous or homozygous variants in NOD2 and patients with wild type NOD2 genotype. Although serum LBP levels have been shown to correlate positively with the state of activity of the disease, TNF-α production did not show a clear correlation with either LBP or sCD14 levels in plasma. Moreover, no clear correlation was seen between TNF-α production and activity indices in either CD or UC. CONCLUSION Peripheral monocytes from CD express higher basal and stimulated TNF-α than controls, regardless of NOD2 genotype and without a clear correlation with disease activity.
Collapse
|
122
|
Luissint AC, Parkos CA, Nusrat A. Inflammation and the Intestinal Barrier: Leukocyte-Epithelial Cell Interactions, Cell Junction Remodeling, and Mucosal Repair. Gastroenterology 2016; 151:616-32. [PMID: 27436072 PMCID: PMC5317033 DOI: 10.1053/j.gastro.2016.07.008] [Citation(s) in RCA: 390] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 06/13/2016] [Accepted: 07/06/2016] [Indexed: 02/06/2023]
Abstract
The intestinal tract is lined by a single layer of columnar epithelial cells that forms a dynamic, permeable barrier allowing for selective absorption of nutrients, while restricting access to pathogens and food-borne antigens. Precise regulation of epithelial barrier function is therefore required for maintaining mucosal homeostasis and depends, in part, on barrier-forming elements within the epithelium and a balance between pro- and anti-inflammatory factors in the mucosa. Pathologic states, such as inflammatory bowel disease, are associated with a leaky epithelial barrier, resulting in excessive exposure to microbial antigens, recruitment of leukocytes, release of soluble mediators, and ultimately mucosal damage. An inflammatory microenvironment affects epithelial barrier properties and mucosal homeostasis by altering the structure and function of epithelial intercellular junctions through direct and indirect mechanisms. We review our current understanding of complex interactions between the intestinal epithelium and immune cells, with a focus on pathologic mucosal inflammation and mechanisms of epithelial repair. We discuss leukocyte-epithelial interactions, as well as inflammatory mediators that affect the epithelial barrier and mucosal repair. Increased knowledge of communication networks between the epithelium and immune system will lead to tissue-specific strategies for treating pathologic intestinal inflammation.
Collapse
Affiliation(s)
- Anny-Claude Luissint
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Charles A Parkos
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Asma Nusrat
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan.
| |
Collapse
|
123
|
Kogut MH, Arsenault RJ. Editorial: Gut Health: The New Paradigm in Food Animal Production. Front Vet Sci 2016; 3:71. [PMID: 27630994 PMCID: PMC5005397 DOI: 10.3389/fvets.2016.00071] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 08/18/2016] [Indexed: 11/13/2022] Open
Affiliation(s)
- Michael H Kogut
- USDA Agricultural Research Service , College Station, TX , USA
| | | |
Collapse
|
124
|
Abstract
Background Understanding of the gut-liver axis is important for the up-to-date management of liver cirrhosis, and changes of intestinal functions form the core of this interesting research field. Summary Most investigators noted small intestinal dysmotility in their patients with liver cirrhosis. Marked changes in the contraction pattern were observed in early manometric studies. The orocecal transit time, particularly small intestinal transit, has generally been reported to be prolonged, which has been demonstrated in multiple investigations to be related to the severity of the liver disease (e.g., Child-Pugh class), the presence of small intestinal bacterial overgrowth (SIBO) and hepatic encephalopathy (HE) as well as a history of spontaneous bacterial peritonitis. Bacteriologically proven SIBO in proximal jejunal aspiration has been reported to be present in up to 59% of cirrhotic patients and is associated with systemic endotoxemia. Clinical and experimental studies suggest that delayed small bowel transit in liver cirrhosis may lead to SIBO, which could contribute to the symptoms of abdominal pain and diarrhea. In addition to autonomic neuropathy, metabolic derangements and diabetic state, SIBO itself may delay intestinal transit in cirrhotic patients. Several studies, both from the West and the East, have shown that the gut microbiota is altered in cirrhotic patients and particularly those with HE. Further, a quantitative change in Bacteroides/Firmicutes ratio, with a prevalence of potentially pathogenic bacteria (e.g., Enterobacteriaceae) and reduction in specific commensals (e.g., Lachnospiraceae), has been described. Structural and functional changes in the intestinal mucosa that contribute to increases in intestinal permeability for bacteria and their products have been observed in patients with liver cirrhosis, which is considered as an important pathogenetic factor for several complications. The mechanism of intestinal barrier dysfunction in cirrhosis is multifactorial, including alcohol, portal hypertension (vascular congestion and dysregulation), endotoxemia, SIBO, local inflammation and, most likely, immunological factors and medications. Key Messages This review summarizes major achievements regarding intestinal dysfunction in cirrhosis for future gastroenterology research. The question of whether this intestinal barrier dysfunction is accompanied and/or at least partly caused by structural and functional changes in the epithelial tight junction proteins is as yet unsolved. Development of new strategies to modulate gut-liver interaction is urgently needed.
Collapse
Affiliation(s)
- Hiroshi Fukui
- Department of Gastroenterology, Endocrinology and Metabolism, Nara Medical University, Kashihara, Japan
| | - Reiner Wiest
- Department of Gastroenterology, University Hospital of Visceral Surgery and Medicine, Bern, Switzerland
| |
Collapse
|
125
|
Fukui H, Wiest R. Changes of Intestinal Functions in Liver Cirrhosis. Inflamm Intest Dis 2016; 1:24-40. [PMID: 29922655 PMCID: PMC5988129 DOI: 10.1159/000444436] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2016] [Accepted: 02/04/2016] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Understanding of the gut-liver axis is important for the up-to-date management of liver cirrhosis, and changes of intestinal functions form the core of this interesting research field. SUMMARY Most investigators noted small intestinal dysmotility in their patients with liver cirrhosis. Marked changes in the contraction pattern were observed in early manometric studies. The orocecal transit time, particularly small intestinal transit, has generally been reported to be prolonged, which has been demonstrated in multiple investigations to be related to the severity of the liver disease (e.g., Child-Pugh class), the presence of small intestinal bacterial overgrowth (SIBO) and hepatic encephalopathy (HE) as well as a history of spontaneous bacterial peritonitis. Bacteriologically proven SIBO in proximal jejunal aspiration has been reported to be present in up to 59% of cirrhotic patients and is associated with systemic endotoxemia. Clinical and experimental studies suggest that delayed small bowel transit in liver cirrhosis may lead to SIBO, which could contribute to the symptoms of abdominal pain and diarrhea. In addition to autonomic neuropathy, metabolic derangements and diabetic state, SIBO itself may delay intestinal transit in cirrhotic patients. Several studies, both from the West and the East, have shown that the gut microbiota is altered in cirrhotic patients and particularly those with HE. Further, a quantitative change in Bacteroides/Firmicutes ratio, with a prevalence of potentially pathogenic bacteria (e.g., Enterobacteriaceae) and reduction in specific commensals (e.g., Lachnospiraceae), has been described. Structural and functional changes in the intestinal mucosa that contribute to increases in intestinal permeability for bacteria and their products have been observed in patients with liver cirrhosis, which is considered as an important pathogenetic factor for several complications. The mechanism of intestinal barrier dysfunction in cirrhosis is multifactorial, including alcohol, portal hypertension (vascular congestion and dysregulation), endotoxemia, SIBO, local inflammation and, most likely, immunological factors and medications. KEY MESSAGES This review summarizes major achievements regarding intestinal dysfunction in cirrhosis for future gastroenterology research. The question of whether this intestinal barrier dysfunction is accompanied and/or at least partly caused by structural and functional changes in the epithelial tight junction proteins is as yet unsolved. Development of new strategies to modulate gut-liver interaction is urgently needed.
Collapse
Affiliation(s)
- Hiroshi Fukui
- Department of Gastroenterology, Endocrinology and Metabolism, Nara Medical University, Kashihara, Japan
| | - Reiner Wiest
- Department of Gastroenterology, University Hospital of Visceral Surgery and Medicine, Bern, Switzerland
| |
Collapse
|
126
|
Abrupt suspension of probiotics administration may increase host pathogen susceptibility by inducing gut dysbiosis. Sci Rep 2016; 6:23214. [PMID: 26983596 PMCID: PMC4794715 DOI: 10.1038/srep23214] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 02/29/2016] [Indexed: 01/14/2023] Open
Abstract
In this study, we investigated the risk associated with suspension of probiotics administration in tilapia, an animal model that may mimic immune-compromised conditions in humans. Tilapias were fed for 14 days using a probiotics-supplemented diet, followed by a three-day suspension of probiotics treatment and a subsequent challenge by Aeromonas hydrophila. Unexpectedly, the suspension of a probiotic strain Lactobacillus plantarum JCM1149 significantly triggered susceptibility of the host to A. hydrophila. We further observed that suspension of JCM1149 resulted in host gut microbiota dysbiosis and the subsequent disorder in the intestinal metabolites (bile acids, amino acids, and glucose) and damage in the intestinal epithelium, giving rise to a condition similar to antibiotics-induced gut dysbiosis, which collectively impaired tilapia’s gut health and resistance to pathogenic challenges. Additionally, we determined that JCM1149 adhered relatively poorly to tilapia intestinal mucosa and was rapidly released from the gastrointestinal tract (GIT) after suspension, with the rapid loss of probiotic strain probably being the direct cause of gut dysbiosis. Finally, three other probiotic Lactobacillus strains with low intestinal mucosa binding activity showed similar rapid loss phenotype following administration suspension, and induced higher host susceptibility to infection, indicating that the risk is a generic phenomenon in Lactobacillus.
Collapse
|
127
|
Abstract
Background Understanding of the gut-liver axis is important for the up-to-date management of liver cirrhosis, and changes of intestinal functions form the core of this interesting research field. Summary Most investigators noted small intestinal dysmotility in their patients with liver cirrhosis. Marked changes in the contraction pattern were observed in early manometric studies. The orocecal transit time, particularly small intestinal transit, has generally been reported to be prolonged, which has been demonstrated in multiple investigations to be related to the severity of the liver disease (e.g., Child-Pugh class), the presence of small intestinal bacterial overgrowth (SIBO) and hepatic encephalopathy (HE) as well as a history of spontaneous bacterial peritonitis. Bacteriologically proven SIBO in proximal jejunal aspiration has been reported to be present in up to 59% of cirrhotic patients and is associated with systemic endotoxemia. Clinical and experimental studies suggest that delayed small bowel transit in liver cirrhosis may lead to SIBO, which could contribute to the symptoms of abdominal pain and diarrhea. In addition to autonomic neuropathy, metabolic derangements and diabetic state, SIBO itself may delay intestinal transit in cirrhotic patients. Several studies, both from the West and the East, have shown that the gut microbiota is altered in cirrhotic patients and particularly those with HE. Further, a quantitative change in Bacteroides/Firmicutes ratio, with a prevalence of potentially pathogenic bacteria (e.g., Enterobacteriaceae) and reduction in specific commensals (e.g., Lachnospiraceae), has been described. Structural and functional changes in the intestinal mucosa that contribute to increases in intestinal permeability for bacteria and their products have been observed in patients with liver cirrhosis, which is considered as an important pathogenetic factor for several complications. The mechanism of intestinal barrier dysfunction in cirrhosis is multifactorial, including alcohol, portal hypertension (vascular congestion and dysregulation), endotoxemia, SIBO, local inflammation and, most likely, immunological factors and medications. Key Messages This review summarizes major achievements regarding intestinal dysfunction in cirrhosis for future gastroenterology research. The question of whether this intestinal barrier dysfunction is accompanied and/or at least partly caused by structural and functional changes in the epithelial tight junction proteins is as yet unsolved. Development of new strategies to modulate gut-liver interaction is urgently needed.
Collapse
Affiliation(s)
- Hiroshi Fukui
- Department of Gastroenterology, Endocrinology and Metabolism, Nara Medical University, Kashihara, Japan
| | - Reiner Wiest
- Department of Gastroenterology, University Hospital of Visceral Surgery and Medicine, Bern, Switzerland
| |
Collapse
|
128
|
Putignani L, Del Chierico F, Vernocchi P, Cicala M, Cucchiara S, Dallapiccola B. Gut Microbiota Dysbiosis as Risk and Premorbid Factors of IBD and IBS Along the Childhood-Adulthood Transition. Inflamm Bowel Dis 2016; 22:487-504. [PMID: 26588090 DOI: 10.1097/mib.0000000000000602] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Gastrointestinal disorders, although clinically heterogeneous, share pathogenic mechanisms, including genetic susceptibility, impaired gut barrier function, altered microbiota, and environmental triggers (infections, social and behavioral factors, epigenetic control, and diet). Gut microbiota has been studied for inflammatory bowel disease (IBD) and irritable bowel syndrome (IBS) in either children or adults, while modifiable gut microbiota features, acting as risk and premorbid factors along the childhood-adulthood transition, have not been thoroughly investigated so far. Indeed, the relationship between variations of the entire host/microbiota/environmental scenario and clinical phenotypes is still not fully understood. In this respect, tracking gut dysbiosis grading may help deciphering host phenotype-genotype associations and microbiota shifts in an integrated top-down omics-based approach within large-scale pediatric and adult case-control cohorts. Large-scale gut microbiota signatures and host inflammation patterns may be integrated with dietary habits, under genetic and epigenetic constraints, providing gut dysbiosis profiles acting as risk predictors of IBD or IBS in preclinical cases. Tracking dysbiosis supports new personalized/stratified IBD and IBS prevention programmes, generating Decision Support System tools. They include (1) high risk or flare-up recurrence -omics-based dysbiosis profiles; (2) microbial and molecular biomarkers of health and disease; (3) -omics-based pipelines for laboratory medicine diagnostics; (4) health apps for self-management of score-based dietary profiles, which can be shared with clinicians for nutritional habit and lifestyle amendment; (5) -omics profiling data warehousing and public repositories for IBD and IBS profile consultation. Dysbiosis-related indexes can represent novel laboratory and clinical medicine tools preventing or postponing the disease, finally interfering with its natural history.
Collapse
Affiliation(s)
- Lorenza Putignani
- *Unit of Parasitology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy; †Unit of Metagenomics, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy; ‡Gastroenterology Unit, University Campus Bio-Medico of Rome, Rome, Italy; §Department of Pediatrics, Center for Pediatric Inflammatory Bowel Disease, University of Rome "La Sapienza," Rome, Italy; and ‖Scientific Directorate, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | | | | | | | | | | |
Collapse
|
129
|
The effect of hydrolysates of proteins from rice milk on the physiological response of enterocytes and on the adhesion of bacteria from healthy and allergic people - an in vitro study. Cent Eur J Immunol 2016; 41:363-375. [PMID: 28450799 PMCID: PMC5382882 DOI: 10.5114/ceji.2016.65135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 04/04/2016] [Indexed: 11/17/2022] Open
Abstract
Designing an optimal diet requires knowledge of the biological activity of food products, particularly in relation to people with food allergies. The hypothesis, which constitutes the basis of this thesis, states that the peptides and glycopeptides released from proteins by enzymatic hydrolysis are able to change the quantity and quality of the human gastrointestinal ecosystem. Such substrates may interfere with adhesion to the intestinal epithelium microbiota and alter enterocytic metabolic activity. The aim of this study was to determine the effect of protein hydrolysates from rice milk substitute on gut epithelial cells and the intestinal microbiota of healthy people and ones suffering from an allergy to milk. The following experimental work applied systems that reflect the conditions occurring in the gastrointestinal tract.
Collapse
|
130
|
Fan P, Tan Y, Jin K, Lin C, Xia S, Han B, Zhang F, Wu L, Ma X. Supplemental lipoic acid relieves post-weaning diarrhoea by decreasing intestinal permeability in rats. J Anim Physiol Anim Nutr (Berl) 2015; 101:136-146. [PMID: 26717901 DOI: 10.1111/jpn.12427] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 09/30/2015] [Indexed: 12/19/2022]
Abstract
Lipoic acid (LA) is a naturally existing substance which widely distributed in the cellular membranes and cytosol of animal cells. Its intracellular functions include quenching of free radicals and repairing oxidized proteins. The purpose of this study was to evaluate the effects of LA on post-weaning diarrhoea using a rat model. Sixty weaned rats were fed either a basal diet or a LA-supplemented diet, or a zinc oxide (ZnO)-supplemented diet as a positive control. Rats in the LA and ZnO groups had better performance and reduced incidence of diarrhoea (p < 0.05). Both LA and ZnO treatments enhanced intestinal homeostatic and architecture, significantly decreased urinary lactulose to mannitol ratios (p < 0.05) and increased the expression of the intestinal mucosal tight junction proteins occludin (OCLN) and zonula occludens protein-1 (ZO-1) (p < 0.05). LA significantly increased the activities of antioxidant enzymes, and reduced glutathione while decreasing the levels of oxidative glutathione and malondialdehyde in the intestinal mucosa (p < 0.05). Furthermore, an in vitro study indicated that supplementation with LA in IEC-6 intestinal epithelial cells significantly enhanced the expression of OCLN and ZO-1 under hydrogen peroxide-induced oxidative stress. Collectively, these results suggest that LA relieves post-weaning diarrhoea by reducing intestinal permeability and improving antioxidant indices.
Collapse
Affiliation(s)
- P Fan
- State Key Lab of Animal Nutrition, China Agricultural University, Beijing, China
| | - Y Tan
- State Key Lab of Animal Nutrition, China Agricultural University, Beijing, China
| | - K Jin
- State Key Lab of Animal Nutrition, China Agricultural University, Beijing, China
| | - C Lin
- State Key Lab of Animal Nutrition, China Agricultural University, Beijing, China
| | - S Xia
- State Key Lab of Animal Nutrition, China Agricultural University, Beijing, China
| | - B Han
- State Key Lab of Animal Nutrition, China Agricultural University, Beijing, China
| | - F Zhang
- State Key Lab of Animal Nutrition, China Agricultural University, Beijing, China
| | - L Wu
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Central Laboratory, Peking University Cancer Hospital & Institute, Beijing, China
| | - X Ma
- State Key Lab of Animal Nutrition, China Agricultural University, Beijing, China.,Department of Internal Medicine, Center for Autophagy Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
131
|
Duodenojejunal Bypass Leads to Altered Gut Microbiota and Strengthened Epithelial Barriers in Rats. Obes Surg 2015; 26:1576-83. [DOI: 10.1007/s11695-015-1968-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
132
|
Huang CY, Yu LCH. Pathophysiological mechanisms of death resistance in colorectal carcinoma. World J Gastroenterol 2015; 21:11777-11792. [PMID: 26557002 PMCID: PMC4631976 DOI: 10.3748/wjg.v21.i41.11777] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 06/18/2015] [Accepted: 08/31/2015] [Indexed: 02/06/2023] Open
Abstract
Colon cancers develop adaptive mechanisms to survive under extreme conditions and display hallmarks of unlimited proliferation and resistance to cell death. The deregulation of cell death is a key factor that contributes to chemoresistance in tumors. In a physiological context, balance between cell proliferation and death, and protection against cell damage are fundamental processes for maintaining gut epithelial homeostasis. The mechanisms underlying anti-death cytoprotection and tumor resistance often bear common pathways, and although distinguishing them would be a challenge, it would also provide an opportunity to develop advanced anti-cancer therapeutics. This review will outline cell death pathways (i.e., apoptosis, necrosis, and necroptosis), and discuss cytoprotective strategies in normal intestinal epithelium and death resistance mechanisms of colon tumor. In colorectal cancers, the intracellular mechanisms of death resistance include the direct alteration of apoptotic and necroptotic machinery and the upstream events modulating death effectors such as tumor suppressor gene inactivation and pro-survival signaling pathways. The autocrine, paracrine and exogenous factors within a tumor microenvironment can also instigate resistance against apoptotic and necroptotic cell death in colon cancers through changes in receptor signaling or transporter uptake. The roles of cyclooxygenase-2/prostaglandin E2, growth factors, glucose, and bacterial lipopolysaccharides in colorectal cancer will be highlighted. Targeting anti-death pathways in the colon cancer tissue might be a promising approach outside of anti-proliferation and anti-angiogenesis strategies for developing novel drugs to treat refractory tumors.
Collapse
|
133
|
Kelly JR, Kennedy PJ, Cryan JF, Dinan TG, Clarke G, Hyland NP. Breaking down the barriers: the gut microbiome, intestinal permeability and stress-related psychiatric disorders. Front Cell Neurosci 2015; 9:392. [PMID: 26528128 PMCID: PMC4604320 DOI: 10.3389/fncel.2015.00392] [Citation(s) in RCA: 700] [Impact Index Per Article: 70.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 09/21/2015] [Indexed: 12/12/2022] Open
Abstract
The emerging links between our gut microbiome and the central nervous system (CNS) are regarded as a paradigm shift in neuroscience with possible implications for not only understanding the pathophysiology of stress-related psychiatric disorders, but also their treatment. Thus the gut microbiome and its influence on host barrier function is positioned to be a critical node within the brain-gut axis. Mounting preclinical evidence broadly suggests that the gut microbiota can modulate brain development, function and behavior by immune, endocrine and neural pathways of the brain-gut-microbiota axis. Detailed mechanistic insights explaining these specific interactions are currently underdeveloped. However, the concept that a "leaky gut" may facilitate communication between the microbiota and these key signaling pathways has gained traction. Deficits in intestinal permeability may underpin the chronic low-grade inflammation observed in disorders such as depression and the gut microbiome plays a critical role in regulating intestinal permeability. In this review we will discuss the possible role played by the gut microbiota in maintaining intestinal barrier function and the CNS consequences when it becomes disrupted. We will draw on both clinical and preclinical evidence to support this concept as well as the key features of the gut microbiota which are necessary for normal intestinal barrier function.
Collapse
Affiliation(s)
- John R Kelly
- Laboratory of Neurogastroenterology, APC Microbiome Institute, University College Cork Cork, Ireland ; Department of Psychiatry and Neurobehavioural Science, University College Cork Cork, Ireland
| | - Paul J Kennedy
- Laboratory of Neurogastroenterology, APC Microbiome Institute, University College Cork Cork, Ireland
| | - John F Cryan
- Laboratory of Neurogastroenterology, APC Microbiome Institute, University College Cork Cork, Ireland ; Department of Anatomy and Neuroscience, University College Cork Cork, Ireland
| | - Timothy G Dinan
- Laboratory of Neurogastroenterology, APC Microbiome Institute, University College Cork Cork, Ireland ; Department of Psychiatry and Neurobehavioural Science, University College Cork Cork, Ireland
| | - Gerard Clarke
- Laboratory of Neurogastroenterology, APC Microbiome Institute, University College Cork Cork, Ireland ; Department of Psychiatry and Neurobehavioural Science, University College Cork Cork, Ireland
| | - Niall P Hyland
- Laboratory of Neurogastroenterology, APC Microbiome Institute, University College Cork Cork, Ireland ; Department of Pharmacology and Therapeutics, University College Cork Cork, Ireland
| |
Collapse
|
134
|
Kelly JR, Kennedy PJ, Cryan JF, Dinan TG, Clarke G, Hyland NP. Breaking down the barriers: the gut microbiome, intestinal permeability and stress-related psychiatric disorders. Front Cell Neurosci 2015. [DOI: 10.3389/fncel.2015.00392 order by 1-- -] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
|
135
|
Kelly JR, Kennedy PJ, Cryan JF, Dinan TG, Clarke G, Hyland NP. Breaking down the barriers: the gut microbiome, intestinal permeability and stress-related psychiatric disorders. Front Cell Neurosci 2015. [DOI: 10.3389/fncel.2015.00392 order by 8029-- -] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
|
136
|
Kelly JR, Kennedy PJ, Cryan JF, Dinan TG, Clarke G, Hyland NP. Breaking down the barriers: the gut microbiome, intestinal permeability and stress-related psychiatric disorders. Front Cell Neurosci 2015. [DOI: 10.3389/fncel.2015.00392 order by 8029-- #] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
|
137
|
Kelly JR, Kennedy PJ, Cryan JF, Dinan TG, Clarke G, Hyland NP. Breaking down the barriers: the gut microbiome, intestinal permeability and stress-related psychiatric disorders. Front Cell Neurosci 2015. [DOI: 10.3389/fncel.2015.00392 order by 1-- gadu] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
|
138
|
Kelly JR, Kennedy PJ, Cryan JF, Dinan TG, Clarke G, Hyland NP. Breaking down the barriers: the gut microbiome, intestinal permeability and stress-related psychiatric disorders. Front Cell Neurosci 2015. [DOI: 10.3389/fncel.2015.00392 order by 8029-- awyx] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
|
139
|
Kelly JR, Kennedy PJ, Cryan JF, Dinan TG, Clarke G, Hyland NP. Breaking down the barriers: the gut microbiome, intestinal permeability and stress-related psychiatric disorders. Front Cell Neurosci 2015. [DOI: 10.3389/fncel.2015.00392 order by 1-- #] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
|
140
|
Kelly JR, Kennedy PJ, Cryan JF, Dinan TG, Clarke G, Hyland NP. Breaking down the barriers: the gut microbiome, intestinal permeability and stress-related psychiatric disorders. Front Cell Neurosci 2015. [DOI: 10.3389/fncel.2015.00392 and 1880=1880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
|
141
|
Interactions of Salmonella enterica subspecies enterica serovar Typhimurium with gut bacteria. Anaerobe 2015; 33:90-7. [DOI: 10.1016/j.anaerobe.2015.02.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 02/23/2015] [Accepted: 02/25/2015] [Indexed: 11/19/2022]
|
142
|
High-fat diet promotes neuronal loss in the myenteric plexus of the large intestine in mice. Dig Dis Sci 2015; 60:841-9. [PMID: 25330870 DOI: 10.1007/s10620-014-3402-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Accepted: 10/11/2014] [Indexed: 12/28/2022]
Abstract
BACKGROUND Obesity is considered a risk factor for other chronic diseases, and diets rich in lipids can cause alterations in the intestinal functions. AIM The aim of this study was to investigate the effects of a high-fat diet (HFD) on the myenteric plexus of the large intestine in mice. METHODS Swiss mice were distributed into four groups: Control animals fed standard chow for 8 and 17 weeks (C8 and C17 groups) and hyperlipidic animals fed HFD for 8 and 17 weeks (Ob8 and Ob17 groups). Immunofluorescence was performed in the large intestine for the morphologic and quantitative analysis of neuronal populations. RESULTS Animals in the Ob17 group exhibited increased body weight and visceral fat gain compared with the C17 group. The intestinal area was also reduced in the two Ob groups. In the proximal colon, the Ob17 group exhibited 16.1 % reduction of the general neuronal density and 33 % reduction of the VIP-immunoreactive (IR) subpopulation. The general neuronal density in the distal colon was reduced by 45 % in the Ob17 group, and the nNOS-IR density was reduced by 35 %. The morphometry of neuronal cell bodies in the Ob17 group exhibited a reduction of the neuronal area of all of the neuronal populations studied in the proximal colon, with a reduction of the subpopulations of nNOS-IR and VIP-IR neurons in the distal colon. CONCLUSIONS The HFD caused neuronal loss in the myenteric plexus, and nitrergic neurons were more resilient. The changes were more pronounced in the distal colon after 17 weeks.
Collapse
|
143
|
Trolox and ascorbic acid reduce direct and indirect oxidative stress in the IPEC-J2 cells, an in vitro model for the porcine gastrointestinal tract. PLoS One 2015; 10:e0120485. [PMID: 25745867 PMCID: PMC4351986 DOI: 10.1371/journal.pone.0120485] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 01/23/2015] [Indexed: 12/21/2022] Open
Abstract
Oxidative stress in the small intestinal epithelium is a major cause of barrier malfunction and failure to regenerate. This study presents a functional in vitro model using the porcine small intestinal epithelial cell line IPEC-J2 to examine the effects of oxidative stress and to estimate the antioxidant and regenerative potential of Trolox, ascorbic acid and glutathione monoethyl ester. Hydrogen peroxide and diethyl maleate affected the tight junction (zona occludens-1) distribution, significantly increased intracellular oxidative stress (CM-H2DCFDA) and decreased the monolayer integrity (transepithelial electrical resistance and FD-4 permeability), viability (neutral red) and wound healing capacity (scratch assay). Trolox (2 mM) and 1 mM ascorbic acid pre-treatment significantly reduced intracellular oxidative stress, increased wound healing capacity and reduced FD-4 permeability in oxidatively stressed IPEC-J2 cell monolayers. All antioxidant pre-treatments increased transepithelial electrical resistance and viability only in diethyl maleate-treated cells. Glutathione monoethyl ester (10 mM) pre-treatment significantly decreased intracellular oxidative stress and monolayer permeability only in diethyl maleate-treated cells. These data demonstrate that the IPEC-J2 oxidative stress model is a valuable tool to screen antioxidants before validation in piglets.
Collapse
|
144
|
Keku TO, Dulal S, Deveaux A, Jovov B, Han X. The gastrointestinal microbiota and colorectal cancer. Am J Physiol Gastrointest Liver Physiol 2015; 308:G351-63. [PMID: 25540232 PMCID: PMC4346754 DOI: 10.1152/ajpgi.00360.2012] [Citation(s) in RCA: 156] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The human gut is home to a complex and diverse microbiota that contributes to the overall homeostasis of the host. Increasingly, the intestinal microbiota is recognized as an important player in human illness such as colorectal cancer (CRC), inflammatory bowel diseases, and obesity. CRC in itself is one of the major causes of cancer mortality in the Western world. The mechanisms by which bacteria contribute to CRC are complex and not fully understood, but increasing evidence suggests a link between the intestinal microbiota and CRC as well as diet and inflammation, which are believed to play a role in carcinogenesis. It is thought that the gut microbiota interact with dietary factors to promote chronic inflammation and CRC through direct influence on host cell physiology, cellular homeostasis, energy regulation, and/or metabolism of xenobiotics. This review provides an overview on the role of commensal gut microbiota in the development of human CRC and explores its association with diet and inflammation.
Collapse
Affiliation(s)
- Temitope O. Keku
- 1Department of Medicine, School of Medicine, University of North Carolina, Chapel Hill, North Carolina; ,2Center for Gastrointestinal Biology and Disease, School of Medicine, University of North Carolina, Chapel Hill, North Carolina; and
| | - Santosh Dulal
- 1Department of Medicine, School of Medicine, University of North Carolina, Chapel Hill, North Carolina; ,2Center for Gastrointestinal Biology and Disease, School of Medicine, University of North Carolina, Chapel Hill, North Carolina; and
| | - April Deveaux
- 1Department of Medicine, School of Medicine, University of North Carolina, Chapel Hill, North Carolina; ,2Center for Gastrointestinal Biology and Disease, School of Medicine, University of North Carolina, Chapel Hill, North Carolina; and
| | - Biljana Jovov
- 1Department of Medicine, School of Medicine, University of North Carolina, Chapel Hill, North Carolina; ,2Center for Gastrointestinal Biology and Disease, School of Medicine, University of North Carolina, Chapel Hill, North Carolina; and
| | - Xuesong Han
- 3Surveillance and Health Services Research, American Cancer Society, Atlanta, Georgia
| |
Collapse
|
145
|
Rieger J, Janczyk P, Hünigen H, Neumann K, Plendl J. Intraepithelial lymphocyte numbers and histomorphological parameters in the porcine gut after Enterococcus faecium NCIMB 10415 feeding in a Salmonella Typhimurium challenge. Vet Immunol Immunopathol 2015; 164:40-50. [DOI: 10.1016/j.vetimm.2014.12.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Revised: 12/07/2014] [Accepted: 12/31/2014] [Indexed: 02/08/2023]
|
146
|
Reshaping faecal gut microbiota composition by the intake of trans-resveratrol and quercetin in high-fat sucrose diet-fed rats. J Nutr Biochem 2015; 26:651-60. [PMID: 25762527 DOI: 10.1016/j.jnutbio.2015.01.002] [Citation(s) in RCA: 338] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Revised: 11/25/2014] [Accepted: 01/07/2015] [Indexed: 12/17/2022]
Abstract
Diet-induced obesity is associated to an imbalance in the normal gut microbiota composition. Resveratrol and quercetin, widely known for their health beneficial properties, have low bioavailability, and when they reach the colon, they are targets of the gut microbial ecosystem. Hence, the use of these molecules in obesity might be considered as a potential strategy to modulate intestinal bacterial composition. The purpose of this study was to determine whether trans-resveratrol and quercetin administration could counteract gut microbiota dysbiosis produced by high-fat sucrose diet (HFS) and, in turn, improve gut health. Wistar rats were randomised into four groups fed an HFS diet supplemented or not with trans-resveratrol [15 mg/kg body weight (BW)/day], quercetin (30 mg/kg BW/day) or a combination of both polyphenols at those doses. Administration of both polyphenols together prevented body weight gain and reduced serum insulin levels. Moreover, individual supplementation of trans-resveratrol and quercetin effectively reduced serum insulin levels and insulin resistance. Quercetin supplementation generated a great impact on gut microbiota composition at different taxonomic levels, attenuating Firmicutes/Bacteroidetes ratio and inhibiting the growth of bacterial species previously associated to diet-induced obesity (Erysipelotrichaceae, Bacillus, Eubacterium cylindroides). Overall, the administration of quercetin was found to be effective in lessening HFS-diet-induced gut microbiota dysbiosis. In contrast, trans-resveratrol supplementation alone or in combination with quercetin scarcely modified the profile of gut bacteria but acted at the intestinal level, altering the mRNA expression of tight-junction proteins and inflammation-associated genes.
Collapse
|
147
|
Yu LCH. Commensal bacterial internalization by epithelial cells: An alternative portal for gut leakiness. Tissue Barriers 2015; 3:e1008895. [PMID: 26451337 DOI: 10.1080/21688370.2015.1008895] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 01/07/2015] [Accepted: 01/14/2015] [Indexed: 02/06/2023] Open
Abstract
Co-existing paracellular and transcellular barrier defect in intestinal epithelium was documented in inflammatory bowel disease, celiac disease, and intestinal obstruction. Mechanisms regarding tight junction disruption have been extensively studied; however, limited progress has been made in research on bacterial transcytosis. Densely packed brush border (BB), with cholesterol-based lipid rafts in the intermicrovillous membrane invagination, serves as an ultrastructural barrier to prevent direct contact of luminal microbes with the cellular soma. Evidence in in vitro epithelial cell cultures and in vivo animal models of bowel obstruction and antibiotic-resistant bacterial infection had indicated that nonpathogenic, noninvasive enteric bacteria may hijack the lipid raft-mediated endocytic pathways. Our studies have shown that low dose interferon-gamma (IFNγ) causes long myosin light chain kinase (MLCK)-dependent terminal web (TW) contraction and BB fanning, allowing bacteria to pass through the consequently widened intermicrovillous cleft to be endocytosed via caveolin-associated lipid rafts. Activation of intracellular innate immune receptors by bacteria-containing endosomes may further induce inflammatory and oxidative stress, leading to secondary tight junction damage. The finding of bacterial internalization preceding tight junction damage suggests that abnormal bacterial uptake by epithelial cells may contribute to the initiation or relapse of chronic intestinal inflammation.
Collapse
Affiliation(s)
- Linda Chia-Hui Yu
- Graduate Institute of Physiology; National Taiwan University ; Taipei, Taiwan, Republic of China
| |
Collapse
|
148
|
Kuo WT, Lee TC, Yang HY, Chen CY, Au YC, Lu YZ, Wu LL, Wei SC, Ni YH, Lin BR, Chen Y, Tsai YH, Kung JT, Sheu F, Lin LW, Yu LCH. LPS receptor subunits have antagonistic roles in epithelial apoptosis and colonic carcinogenesis. Cell Death Differ 2015; 22:1590-604. [PMID: 25633197 DOI: 10.1038/cdd.2014.240] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 11/24/2014] [Accepted: 12/22/2014] [Indexed: 12/15/2022] Open
Abstract
Colorectal carcinoma (CRC) is characterized by unlimited proliferation and suppression of apoptosis, selective advantages for tumor survival, and chemoresistance. Lipopolysaccharide (LPS) signaling is involved in both epithelial homeostasis and tumorigenesis, but the relative roles had by LPS receptor subunits CD14 and Toll-like receptor 4 (TLR4) are poorly understood. Our study showed that normal human colonocytes were CD14(+)TLR4(-), whereas cancerous tissues were CD14(+)TLR4(+), by immunofluorescent staining. Using a chemical-induced CRC model, increased epithelial apoptosis and decreased tumor multiplicity and sizes were observed in TLR4-mutant mice compared with wild-type (WT) mice with CD14(+)TLR4(+) colonocytes. WT mice intracolonically administered a TLR4 antagonist displayed tumor reduction associated with enhanced apoptosis in cancerous tissues. Mucosa-associated LPS content was elevated in response to CRC induction. Epithelial apoptosis induced by LPS hypersensitivity in TLR4-mutant mice was prevented by intracolonic administration of neutralizing anti-CD14. Moreover, LPS-induced apoptosis was observed in primary colonic organoid cultures derived from TLR4 mutant but not WT murine crypts. Gene silencing of TLR4 increased cell apoptosis in WT organoids, whereas knockdown of CD14 ablated cell death in TLR4-mutant organoids. In vitro studies showed that LPS challenge caused apoptosis in Caco-2 cells (CD14(+)TLR4(-)) in a CD14-, phosphatidylcholine-specific phospholipase C-, sphingomyelinase-, and protein kinase C-ζ-dependent manner. Conversely, expression of functional but not mutant TLR4 (Asp299Gly, Thr399Ile, and Pro714His) rescued cells from LPS/CD14-induced apoptosis. In summary, CD14-mediated lipid signaling induced epithelial apoptosis, whereas TLR4 antagonistically promoted cell survival and cancer development. Our findings indicate that dysfunction in the CD14/TLR4 antagonism may contribute to normal epithelial transition to carcinogenesis, and provide novel strategies for intervention against colorectal cancer.
Collapse
Affiliation(s)
- W-T Kuo
- Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - T-C Lee
- Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan.,Department of Internal Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| | - H-Y Yang
- Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - C-Y Chen
- Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Y-C Au
- Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Y-Z Lu
- Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - L-L Wu
- Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - S-C Wei
- Department of Internal Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Y-H Ni
- Department of Pediatrics, National Taiwan University College of Medicine, Taipei, Taiwan
| | - B-R Lin
- Department of Surgery, National Taiwan University College of Medicine and Hospital, Taipei, Taiwan
| | - Y Chen
- Department of Surgery, Far Eastern Memorial Hospital, New Taipei, Taiwan.,Department of Chemical Engineering and Material Science, Yuan-Ze University, Tao-Yuan, Taiwan
| | - Y-H Tsai
- Department of Surgery, Far Eastern Memorial Hospital, New Taipei, Taiwan.,Department of Chemical Engineering and Material Science, Yuan-Ze University, Tao-Yuan, Taiwan
| | - J T Kung
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - F Sheu
- Department of Horticulture, National Taiwan University, Taipei, Taiwan
| | - L-W Lin
- Department of Pathology, National Taiwan University Hospital, Yunlin Branch, Yunlin, Taiwan
| | - L C-H Yu
- Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan
| |
Collapse
|
149
|
|
150
|
Understanding host-adherent-invasive Escherichia coli interaction in Crohn's disease: opening up new therapeutic strategies. BIOMED RESEARCH INTERNATIONAL 2014; 2014:567929. [PMID: 25580435 PMCID: PMC4279263 DOI: 10.1155/2014/567929] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 09/16/2014] [Accepted: 09/16/2014] [Indexed: 02/07/2023]
Abstract
A trillion of microorganisms colonize the mammalian intestine. Most of them have coevolved with the host in a symbiotic relationship and some of them have developed strategies to promote their replication in the presence of competing microbiota. Recent evidence suggests that perturbation of the microbial community favors the emergence of opportunistic pathogens, in particular adherent-invasive Escherichia coli (AIEC) that can increase incidence and severity of gut inflammation in the context of Crohn's disease (CD). This review will report the importance of AIEC as triggers of intestinal inflammation, focusing on their impact on epithelial barrier function and stimulation of mucosal inflammation. Beyond manipulation of immune response, restoration of gut microbiota as a new treatment option for CD patients will be discussed.
Collapse
|