1
|
Kovanda L, Hejna M, Du T, Liu Y. Butyrate Derivatives Exhibited Anti-Inflammatory Effects and Enhanced Intestinal Barrier Integrity in Porcine Cell Culture Models. Animals (Basel) 2025; 15:1289. [PMID: 40362102 PMCID: PMC12071038 DOI: 10.3390/ani15091289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 04/25/2025] [Accepted: 04/29/2025] [Indexed: 05/15/2025] Open
Abstract
Butyrate and its derivatives may influence inflammatory status and physiology in a variety of organisms and organ systems. Inflammatory conditions of the gastrointestinal tract, such as post-weaning diarrhea, negatively impact swine. Dietary intervention with butyrate-based compounds should be considered a strategy to improve disease resistance in pigs. We aimed to assess the properties of different forms of butyrate treatments using porcine cell culture experiments. This assessment may inform future in vivo feed experiments designed to determine its potential application of the dietary supplements for pigs. An intestinal porcine enterocyte cell line, IPEC-J2, was seeded at 5 × 103 cells/mL in 96-well plates to confirm cell viability by MTT assay for each dose range used in the current experiments (0, 0.5, 1, 2, 4 mM butyric acid or tributyrin; 0, 1, 2, 4, 8 mM sodium butyrate or monobutyrin). For transepithelial electrical resistance (TEER) analysis, IPEC-J2 was seeded at 5 × 105 cells/mL in 12-well transwell inserts and treated with 5 levels of each butyrate derivative after adherence (n = 5). TEER was measured at 24, 48, and 72 h post-treatment to quantify intestinal barrier integrity of IPEC-J2 monolayers. Butyric acid, sodium butyrate, and monobutyrin significantly increased (p < 0.05) TEER in IPEC-J2 at different time points compared with control. Further, porcine alveolar macrophages (PAMs) were harvested from donor weaned piglets (n = 6) via bronchoalveolar lavage and isolated for primary culture (6 × 105 cells/well, 6-well plates). PAMs were treated with five levels of each butyrate derivative with or without lipopolysaccharide (LPS, 1 μg/mL) challenge. The concentrations of TNF-α and IL-1β in cell culture supernatants were measured by enzyme-linked immunosorbent assay (ELISA). Butyric acid and sodium butyrate treatments reduced the production of TNF-α in LPS-challenged PAMs (linear; p < 0.05). Different butyrate derivatives exerted anti-inflammatory properties and improved intestinal barrier integrity.
Collapse
Affiliation(s)
- Lauren Kovanda
- Department of Animal Science, University of California, Davis, CA 95616, USA; (L.K.); (T.D.)
| | - Monika Hejna
- Department of Biotechnology and Nutrigenomics, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Postępu 36A, 05-552 Jastrzębiec, Poland;
| | - Tina Du
- Department of Animal Science, University of California, Davis, CA 95616, USA; (L.K.); (T.D.)
- Department of Nutrition, University of California, Davis, CA 95616, USA
| | - Yanhong Liu
- Department of Animal Science, University of California, Davis, CA 95616, USA; (L.K.); (T.D.)
| |
Collapse
|
2
|
Abramov VM, Kosarev IV, Machulin AV, Deryusheva EI, Priputnevich TV, Panin AN, Chikileva IO, Abashina TN, Manoyan AM, Akhmetzyanova AA, Blumenkrants DA, Ivanova OE, Papazyan TT, Nikonov IN, Suzina NE, Melnikov VG, Khlebnikov VS, Sakulin VK, Samoilenko VA, Gordeev AB, Sukhikh GT, Uversky VN, Karlyshev AV. Anti- Salmonella Defence and Intestinal Homeostatic Maintenance In Vitro of a Consortium Containing Limosilactobacillus fermentum 3872 and Ligilactobacillus salivarius 7247 Strains in Human, Porcine, and Chicken Enterocytes. Antibiotics (Basel) 2023; 13:30. [PMID: 38247590 PMCID: PMC10812507 DOI: 10.3390/antibiotics13010030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 12/18/2023] [Accepted: 12/22/2023] [Indexed: 01/23/2024] Open
Abstract
Limosilactobacillus fermentum strain 3872 (LF3872) was originally isolated from the breast milk of a healthy woman during lactation and the breastfeeding of a child. Ligilactobacillus salivarius strain 7247 (LS7247) was isolated at the same time from the intestines and reproductive system of a healthy woman. The genomes of these strains contain genes responsible for the production of peptidoglycan-degrading enzymes and factors that increase the permeability of the outer membrane of Gram-negative pathogens. In this work, the anti-Salmonella and intestinal homeostatic features of the LF3872 and LS7247 consortium were studied. A multi-drug resistant (MDR) strain of Salmonella enteritidis (SE) was used in the experiments. The consortium effectively inhibited the adhesion of SE to intact and activated human, porcine, and chicken enterocytes and reduced invasion. The consortium had a bactericidal effect on SE in 6 h of co-culturing. A gene expression analysis of SE showed that the cell-free supernatant (CFS) of the consortium inhibited the expression of virulence genes critical for the colonization of human and animal enterocytes. The CFS stimulated the production of an intestinal homeostatic factor-intestinal alkaline phosphatase (IAP)-in Caco-2 and HT-29 enterocytes. The consortium decreased the production of pro-inflammatory cytokines IL-8, TNF-α, and IL-1β, and TLR4 mRNA expression in human and animal enterocytes. It stimulated the expression of TLR9 in human and porcine enterocytes and stimulated the expression of TLR21 in chicken enterocytes. The consortium also protected the intestinal barrier functions through the increase of transepithelial electrical resistance (TEER) and the inhibition of paracellular permeability in the monolayers of human and animal enterocytes. The results obtained suggest that a LF3872 and LS7247 consortium can be used as an innovative feed additive to reduce the spread of MDR SE among the population and farm animals.
Collapse
Affiliation(s)
- Vyacheslav M. Abramov
- Federal Service for Veterinary and Phytosanitary Surveillance (Rosselkhoznadzor) Federal State Budgetary Institution “The Russian State Center for Animal Feed and Drug Standardization and Quality” (FGBU VGNKI), 123022 Moscow, Russia
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Health, 117997 Moscow, Russia (A.B.G.)
| | - Igor V. Kosarev
- Federal Service for Veterinary and Phytosanitary Surveillance (Rosselkhoznadzor) Federal State Budgetary Institution “The Russian State Center for Animal Feed and Drug Standardization and Quality” (FGBU VGNKI), 123022 Moscow, Russia
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Health, 117997 Moscow, Russia (A.B.G.)
| | - Andrey V. Machulin
- Skryabin Institute of Biochemistry and Physiology of Microorganisms, Federal Research Center “Pushchino Scientific Center for Biological Research of Russian Academy of Science”, Russian Academy of Science, 142290 Pushchino, Russia
| | - Evgenia I. Deryusheva
- Institute for Biological Instrumentation, Federal Research Center “Pushchino Scientific Center for Biological Research of Russian Academy of Science”, Russian Academy of Science, 142290 Pushchino, Russia
| | - Tatiana V. Priputnevich
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Health, 117997 Moscow, Russia (A.B.G.)
| | - Alexander N. Panin
- Federal Service for Veterinary and Phytosanitary Surveillance (Rosselkhoznadzor) Federal State Budgetary Institution “The Russian State Center for Animal Feed and Drug Standardization and Quality” (FGBU VGNKI), 123022 Moscow, Russia
| | - Irina O. Chikileva
- Blokhin National Research Center of Oncology, Ministry of Health RF, 115478 Moscow, Russia
| | - Tatiana N. Abashina
- Skryabin Institute of Biochemistry and Physiology of Microorganisms, Federal Research Center “Pushchino Scientific Center for Biological Research of Russian Academy of Science”, Russian Academy of Science, 142290 Pushchino, Russia
| | - Ashot M. Manoyan
- Federal Service for Veterinary and Phytosanitary Surveillance (Rosselkhoznadzor) Federal State Budgetary Institution “The Russian State Center for Animal Feed and Drug Standardization and Quality” (FGBU VGNKI), 123022 Moscow, Russia
| | - Anna A. Akhmetzyanova
- Federal Service for Veterinary and Phytosanitary Surveillance (Rosselkhoznadzor) Federal State Budgetary Institution “The Russian State Center for Animal Feed and Drug Standardization and Quality” (FGBU VGNKI), 123022 Moscow, Russia
| | - Dmitriy A. Blumenkrants
- Federal Service for Veterinary and Phytosanitary Surveillance (Rosselkhoznadzor) Federal State Budgetary Institution “The Russian State Center for Animal Feed and Drug Standardization and Quality” (FGBU VGNKI), 123022 Moscow, Russia
| | - Olga E. Ivanova
- Federal Service for Veterinary and Phytosanitary Surveillance (Rosselkhoznadzor) Federal State Budgetary Institution “The Russian State Center for Animal Feed and Drug Standardization and Quality” (FGBU VGNKI), 123022 Moscow, Russia
| | | | - Ilia N. Nikonov
- Federal State Educational Institution of Higher Professional Education, Moscow State Academy of Veterinary Medicine and Biotechnology Named after K.I. Skryabin, 109472 Moscow, Russia;
| | - Nataliya E. Suzina
- Skryabin Institute of Biochemistry and Physiology of Microorganisms, Federal Research Center “Pushchino Scientific Center for Biological Research of Russian Academy of Science”, Russian Academy of Science, 142290 Pushchino, Russia
| | - Vyacheslav G. Melnikov
- Gabrichevsky Research Institute for Epidemiology and Microbiology, 125212 Moscow, Russia
| | | | - Vadim K. Sakulin
- Institute of Immunological Engineering, 142380 Lyubuchany, Russia; (V.S.K.); (V.K.S.)
| | - Vladimir A. Samoilenko
- Skryabin Institute of Biochemistry and Physiology of Microorganisms, Federal Research Center “Pushchino Scientific Center for Biological Research of Russian Academy of Science”, Russian Academy of Science, 142290 Pushchino, Russia
| | - Alexey B. Gordeev
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Health, 117997 Moscow, Russia (A.B.G.)
| | - Gennady T. Sukhikh
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Health, 117997 Moscow, Russia (A.B.G.)
| | - Vladimir N. Uversky
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA;
| | - Andrey V. Karlyshev
- Department of Biomolecular Sciences, School of Life Sciences, Chemistry and Pharmacy, Faculty of Health, Science, Social Care and Education, Kingston University London, Kingston upon Thames KT1 2EE, UK
| |
Collapse
|
3
|
Du X, Rodriguez J, Wee J. Dietary Postbiotics Reduce Cytotoxicity and Inflammation Induced by Crystalline Silica in an In Vitro RAW 264.7 Macrophage Model. Foods 2022; 11:foods11060877. [PMID: 35327299 PMCID: PMC8955347 DOI: 10.3390/foods11060877] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 03/14/2022] [Accepted: 03/16/2022] [Indexed: 11/21/2022] Open
Abstract
Crystalline silica (cSiO2) particles are naturally existing environmental toxicants. Exposure to cSiO2 could cause local or systemic inflammation and aggregate inflammation-associated diseases. Dietary postbiotics are reported to possess anti-inflammatory activities; however, their effects on cSiO2-triggered inflammation are unknown. Here, we investigate the impact of postbiotics from Lacticaseibacillus rhamnosus (LGG), Limosilactobacillus reuteri (L.reu), and Bifidobacterium animalis subsp. lactis Bb12 (BB12) on cSiO2-induced cytotoxicity and IL-1 cytokines in vitro using macrophages. The postbiotics used in this study were cell-free fractions of a probiotic growth medium collected at different time points. The in vitro model used was the wild-type murine macrophage RAW 264.7 cell line stably transfected with the inflammasome adapter protein, ASC. Our results indicate that all the postbiotics could reduce cSiO2-induced cytotoxicity in the wild-type and ASC macrophages and the effects were OD-dependent. Following priming with a lipopolysaccharide, cSiO2 treatment resulted in robust inflammasome activation in ASC, as reflected by the IL-1β release. These responses were minimal or absent in the wild-type RAW cells. All the postbiotics decreased the release of IL-1β from ASC; however, only LGG and BB12 reduced the IL-1β secretion from wild-type cells. Only the L.reu postbiotics reduced the IL-1α release from ASC. We conclude that the postbiotics from LGG, BB12, and L.reu can protect macrophages against cSiO2-induced cytotoxicity and suppress IL-1β activation.
Collapse
|
4
|
Pellerin G, Bazinet L, Grenier D. Effect of cranberry juice deacidification on its antibacterial activity against periodontal pathogens and its anti-inflammatory properties in an oral epithelial cell model. Food Funct 2021; 12:10470-10483. [PMID: 34554173 DOI: 10.1039/d1fo01552d] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Cranberries are widely recognized as a functional food that can promote oral health. However, the high concentration of organic acids in cranberry juice can cause tooth enamel erosion. Electrodialysis with bipolar membrane (EDBM) is a process used for the deacidification of cranberry juice. The present study investigated whether the removal of organic acids (0%, 19%, 42%, 60%, and 79%) from cranberry juice by EDBM affects its antibacterial activity against major periodontopathogens as well as its anti-inflammatory properties in an oral epithelial cell model. A deacidification rate ≥60% attenuated the bactericidal effect against planktonic and biofilm-embedded Aggregatibacter actinomycetemcomitans but had no impact on Porphyromonas gingivalis and Fusobacterium nucleatum. Cranberry juice increased the adherence of A. actinomycetemcomitans and P. gingivalis to oral epithelial cells, but reduced the adherence of F. nucleatum by half regardless of the deacidification rate. F. nucleatum produced more hydrogen sulfide when it was exposed to deacidified cranberry juice with a deacidification rate ≥42% compared to the raw beverage. Interestingly, the removal of organic acids from cranberry juice lowered the cytotoxicity of the beverage for oral epithelial cells. Deacidification attenuated the anti-inflammatory effect of cranberry juice in an in vitro oral epithelial cell model. The secretion of IL-6 by lipopolysaccharide (LPS)-stimulated oral epithelial cells exposed to cranberry juice increased proportionally with the deacidification rate. No such effect was observed with respect to the production of IL-8. This study provided evidence that organic acids, just like phenolic compounds, might contribute to the health benefits of cranberry juice against periodontitis.
Collapse
Affiliation(s)
- Geneviève Pellerin
- Institute of Nutrition and Functional Foods (INAF) and Department of Food Sciences, Université Laval, Quebec City, QC, Canada G1V 0A6.,Laboratoire de Transformation Alimentaire et Procédés Électro-Membranaires (LTAPEM, Laboratory of Food Processing and Electromembrane Processes), Université Laval, Quebec City, QC, Canada G1V 0A6.
| | - Laurent Bazinet
- Institute of Nutrition and Functional Foods (INAF) and Department of Food Sciences, Université Laval, Quebec City, QC, Canada G1V 0A6.,Laboratoire de Transformation Alimentaire et Procédés Électro-Membranaires (LTAPEM, Laboratory of Food Processing and Electromembrane Processes), Université Laval, Quebec City, QC, Canada G1V 0A6.
| | - Daniel Grenier
- Oral Ecology Research Group, Faculty of Dentistry, Université Laval, Quebec City, QC, Canada G1V 0A6.
| |
Collapse
|
5
|
Characterization and Cytotoxic Evaluation of Bacteriocins Possessing Antibiofilm Activity Produced by Lactobacillus plantarum SJ33. Int J Pept Res Ther 2021. [DOI: 10.1007/s10989-021-10210-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
6
|
Gupta A, Bansal M, Liyanage R, Upadhyay A, Rath N, Donoghue A, Sun X. Sodium butyrate modulates chicken macrophage proteins essential for Salmonella Enteritidis invasion. PLoS One 2021; 16:e0250296. [PMID: 33909627 PMCID: PMC8081216 DOI: 10.1371/journal.pone.0250296] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 04/02/2021] [Indexed: 12/28/2022] Open
Abstract
Salmonella Enteritidis is an intracellular foodborne pathogen that has developed multiple mechanisms to alter poultry intestinal physiology and infect the gut. Short chain fatty acid butyrate is derived from microbiota metabolic activities, and it maintains gut homeostasis. There is limited understanding on the interaction between S. Enteritidis infection, butyrate, and host intestinal response. To fill this knowledge gap, chicken macrophages (also known as HTC cells) were infected with S. Enteritidis, treated with sodium butyrate, and proteomic analysis was performed. A growth curve assay was conducted to determine sub-inhibitory concentration (SIC, concentration that do not affect bacterial growth compared to control) of sodium butyrate against S. Enteritidis. HTC cells were infected with S. Enteritidis in the presence and absence of SIC of sodium butyrate. The proteins were extracted and analyzed by tandem mass spectrometry. Our results showed that the SIC was 45 mM. Notably, S. Enteritidis-infected HTC cells upregulated macrophage proteins involved in ATP synthesis through oxidative phosphorylation such as ATP synthase subunit alpha (ATP5A1), ATP synthase subunit d, mitochondrial (ATP5PD) and cellular apoptosis such as Cytochrome-c (CYC). Furthermore, sodium butyrate influenced S. Enteritidis-infected HTC cells by reducing the expression of macrophage proteins mediating actin cytoskeletal rearrangements such as WD repeat-containing protein-1 (WDR1), Alpha actinin-1 (ACTN1), Vinculin (VCL) and Protein disulfide isomerase (P4HB) and intracellular S. Enteritidis growth and replication such as V-type proton ATPase catalytic subunit A (ATPV1A). Interestingly, sodium butyrate increased the expression of infected HTC cell protein involving in bacterial killing such as Vimentin (VIM). In conclusion, sodium butyrate modulates the expression of HTC cell proteins essential for S. Enteritidis invasion.
Collapse
Affiliation(s)
- Anamika Gupta
- Department of Poultry Science, University of Arkansas, Fayetteville, Arkansas, United States of America
| | - Mohit Bansal
- Department of Poultry Science, University of Arkansas, Fayetteville, Arkansas, United States of America
| | - Rohana Liyanage
- Department of Chemistry, University of Arkansas, Fayetteville, Arkansas, United States of America
| | - Abhinav Upadhyay
- Department of Animal Science, University of Connecticut, Storrs, Connecticut, United States of America
| | - Narayan Rath
- Poultry Production and Product Safety Research Unit, United States Department of Agriculture-Agriculture Research Station, Fayetteville, Arkansas, United States of America
| | - Annie Donoghue
- Poultry Production and Product Safety Research Unit, United States Department of Agriculture-Agriculture Research Station, Fayetteville, Arkansas, United States of America
| | - Xiaolun Sun
- Department of Poultry Science, University of Arkansas, Fayetteville, Arkansas, United States of America
| |
Collapse
|
7
|
Gupta A, Bansal M, Wagle B, Sun X, Rath N, Donoghue A, Upadhyay A. Sodium Butyrate Reduces Salmonella Enteritidis Infection of Chicken Enterocytes and Expression of Inflammatory Host Genes in vitro. Front Microbiol 2020; 11:553670. [PMID: 33042060 PMCID: PMC7524895 DOI: 10.3389/fmicb.2020.553670] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 08/27/2020] [Indexed: 12/14/2022] Open
Abstract
Salmonella Enteritidis (SE) is a facultative intracellular pathogen that colonizes the chicken gut leading to contamination of carcasses during processing. A reduction in intestinal colonization by SE could result in reduced carcass contamination thereby reducing the risk of illnesses in humans. Short chain fatty acids such as butyrate are microbial metabolites produced in the gut that exert various beneficial effects. However, its effect on SE colonization is not well known. The present study investigated the effect of sub-inhibitory concentrations (SICs) of sodium butyrate on the adhesion and invasion of SE in primary chicken enterocytes and chicken macrophages. In addition, the effect of sodium butyrate on the expression of SE virulence genes and selected inflammatory genes in chicken macrophages challenged with SE were investigated. Based on the growth curve analysis, the two SICs of sodium butyrate that did not reduce SE growth were 22 and 45 mM, respectively. The SICs of sodium butyrate did not affect the viability and proliferation of chicken enterocytes and macrophage cells. The SICs of sodium butyrate reduced SE adhesion by ∼1.7 and 1.8 Log CFU/mL, respectively. The SE invasion was reduced by ∼2 and 2.93 Log CFU/mL, respectively in chicken enterocytes (P < 0.05). Sodium butyrate did not significantly affect the adhesion of SE to chicken macrophages. However, 45 mM sodium butyrate reduced invasion by ∼1.7 Log CFU/mL as compared to control (P < 0.05). Exposure to sodium butyrate did not change the expression of SE genes associated with motility (flgG, prot6E), invasion (invH), type 3 secretion system (sipB, pipB), survival in macrophages (spvB, mgtC), cell wall and membrane integrity (tatA), efflux pump regulator (mrr1) and global virulence regulation (lrp) (P > 0.05). However, a few genes contributing to type-3 secretion system (ssaV, sipA), adherence (sopB), macrophage survival (sodC) and oxidative stress (rpoS) were upregulated by at least twofold. The expression of inflammatory genes (Il1β, Il8, and Mmp9) that are triggered by SE for host colonization was significantly downregulated (at least 25-fold) by sodium butyrate as compared to SE (P < 0.05). The results suggest that sodium butyrate has an anti-inflammatory potential to reduce SE colonization in chickens.
Collapse
Affiliation(s)
- Anamika Gupta
- Department of Poultry Science, University of Arkansas, Fayetteville, AR, United States
| | - Mohit Bansal
- Department of Poultry Science, University of Arkansas, Fayetteville, AR, United States
| | - Basanta Wagle
- Department of Poultry Science, University of Arkansas, Fayetteville, AR, United States
| | - Xiaolun Sun
- Department of Poultry Science, University of Arkansas, Fayetteville, AR, United States
| | - Narayan Rath
- Poultry Production and Product Safety Research Unit, United States Department of Agriculture-Agriculture Research Station, Fayetteville, AR, United States
| | - Annie Donoghue
- Poultry Production and Product Safety Research Unit, United States Department of Agriculture-Agriculture Research Station, Fayetteville, AR, United States
| | - Abhinav Upadhyay
- Department of Animal Science, University of Connecticut, Storrs, CT, United States
| |
Collapse
|
8
|
Pagliai G, Russo E, Niccolai E, Dinu M, Di Pilato V, Magrini A, Bartolucci G, Baldi S, Menicatti M, Giusti B, Marcucci R, Rossolini GM, Casini A, Sofi F, Amedei A. Influence of a 3-month low-calorie Mediterranean diet compared to the vegetarian diet on human gut microbiota and SCFA: the CARDIVEG Study. Eur J Nutr 2020; 59:2011-2024. [PMID: 31292752 DOI: 10.1007/s00394-019-02050-0] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 07/05/2019] [Indexed: 12/27/2022]
Abstract
PURPOSE We evaluated the effect of low-calorie mediterranean (MD) and vegetarian (VD) diets on gut microbiome (GM) composition and short-chain-fatty acids (SCFA) production. METHODS We performed next generation sequencing (NGS) of 16S rRNA and SCFA analysis on fecal samples of 23 overweight omnivores (16 F; 7 M) with low-to-moderate cardiovascular risk. They were randomly assigned to a VD or MD, each lasting 3 months, with a crossover study design. RESULTS Dietary interventions did not produce significant diversity in the GM composition at higher ranks (family and above), neither between nor within MD and VD, but they did it at genus level. MD significantly changed the abundance of Enterorhabdus, Lachnoclostridium and Parabacteroides, while VD significantly affected the abundance of Anaerostipes, Streptococcus, Clostridium sensu stricto, and Odoribacter. Comparison of the mean variation of each SCFA between MD and VD showed an opposite and statistically significant trend for propionic acid (+ 10% vs - 28%, respectively, p = 0.034). In addition, variations of SCFA were negatively correlated with changes of some inflammatory cytokines such as VEGF, MCP-1, IL-17, IP-10 and IL-12, only after MD. Finally, correlation analyses showed a potential relationship-modulated by the two diets-between changes of genera and changes of clinical and biochemical parameters. CONCLUSIONS A short-term dietary intervention with MD or VD does not induce major change in the GM, suggesting that a diet should last longer than 3 months for scratching the microbial resilience. Changes in SCFA production support their role in modulating the inflammatory response, thus mediating the anti-inflammatory and protective properties of MD.
Collapse
Affiliation(s)
- Giuditta Pagliai
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Edda Russo
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Elena Niccolai
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Monica Dinu
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Vincenzo Di Pilato
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Alessandro Magrini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Gianluca Bartolucci
- Department of Neurosciences, Psychology, Drug Research and Child Health Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Viale Pieraccini 6, 50139, Florence, Italy
| | - Simone Baldi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Marta Menicatti
- Department of Neurosciences, Psychology, Drug Research and Child Health Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Viale Pieraccini 6, 50139, Florence, Italy
| | - Betti Giusti
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Atherothrombotic Diseases Unit, Careggi University Hospital, Florence, Italy
| | - Rossella Marcucci
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Atherothrombotic Diseases Unit, Careggi University Hospital, Florence, Italy
| | - Gian Maria Rossolini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Alessandro Casini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Clinical Nutrition Unit, Careggi University Hospital, Florence, Italy
| | - Francesco Sofi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Clinical Nutrition Unit, Careggi University Hospital, Florence, Italy
- IRCCS Don Carlo Gnocchi Foundation, Florence, Italy
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.
- Department of Biomedicine, Careggi University Hospital, Florence, Italy.
| |
Collapse
|
9
|
Abstract
PURPOSE Butyrate is a short-chain fatty acid produced in the intestine. It is controversial whether butyrate is protective or destructive for the intestinal epithelium in the development of diseases like necrotizing enterocolitis (NEC), and its mechanism of action remains unclear. We aimed to determine the effect of butyrate on the intestinal epithelium by studying its effects on intestinal epithelial cells (IEC-18) exposed to injury and in vivo by investigating the effects on the intestine in an experimental model of NEC. METHODS A) In vitro study: Butyrate was given to normal IEC-18 to determine the dose triggering injury. Based on above results, low dose butyrate (1 mM) was given to H2O2-injured cells to determine its effect against inflammation. B) In vivo study: NEC was induced by hypoxia and gavage feeding between postnatal day P5 and P9 (n = 8). Breastfed mice were used as control (n = 7). Butyrate (150 mM) was administered by enema on P6 in NEC (n = 6). Distal ileum was harvested on P9. RESULTS High dose (16 mM) butyrate upregulated inflammatory marker IL-6, while low dose butyrate protected cells from injury by reducing IL-6 expression. Similarly, compared with NEC alone, NEC mice who received butyrate had reduced intestinal damage, reduced IL-6 and NF-ĸB expression, and increased intestinal tight junction marker Claudin-7. CONCLUSION Butyrate has opposite effects depending on the dose administered. Butyrate can protect cells from H2O2-induced injury and can in vivo protect the intestine from NEC. This beneficial effect is because of downregulation of inflammation and enhancement of intestinal barrier.
Collapse
|
10
|
Inhibiting bacterial colonization on catheters: Antibacterial and antibiofilm activities of bacteriocins from Lactobacillus plantarum SJ33. J Glob Antimicrob Resist 2019; 19:85-92. [DOI: 10.1016/j.jgar.2019.02.021] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 02/27/2019] [Accepted: 02/27/2019] [Indexed: 01/08/2023] Open
|
11
|
Nguyen TTT, Fujimura Y, Mimura I, Fujii Y, Nguyen NL, Arakawa K, Morita H. Cultivable butyrate-producing bacteria of elderly Japanese diagnosed with Alzheimer’s disease. J Microbiol 2018; 56:760-771. [DOI: 10.1007/s12275-018-8297-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 07/04/2018] [Accepted: 07/04/2018] [Indexed: 12/16/2022]
|
12
|
Inhibition of tumour and non-tumour cell proliferation by pygidial gland secretions of four ground beetle species (Coleoptera: Carabidae). Biologia (Bratisl) 2018. [DOI: 10.2478/s11756-018-0082-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
13
|
Is cytotoxicity a determinant of the different in vitro and in vivo effects of bioactives? Altern Ther Health Med 2017; 17:453. [PMID: 28882181 PMCID: PMC5590171 DOI: 10.1186/s12906-017-1962-2] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 09/01/2017] [Indexed: 12/12/2022]
Abstract
BACKGROUND Foodstuffs of both plant and animal origin contain a wide range of bioactive compounds. Although human intervention studies are mandatory to assess the health effects of bioactives, the in vitro approach is often used to select the most promising molecules to be studied in vivo. To avoid misleading results, concentration and chemical form, exposure time, and potential cytotoxicity of the tested bioactives should be carefully set prior to any other experiments. METHODS In this study the possible cytotoxicity of different bioactives (docosahexaenoic acid, propionate, cyanidin-3-O-glucoside, protocatechuic acid), was investigated in HepG2 cells using different methods. Bioactives were supplemented to cells at different concentrations within the physiological range in human blood, alone or in combination, considering two different exposure times. RESULTS Reported data clearly evidence that in vitro cytotoxicity is tightly related to the exposure time, and it varies among bioactives, which could exert a cytotoxic effect even at a concentration within the in vivo physiological blood concentration range. Furthermore, co-supplementation of different bioactives can increase the cytotoxic effect. CONCLUSIONS Our results underline the importance of in vitro cytotoxicity screening that should be considered mandatory before performing studies aimed to evaluate the effect of bioactives on other cellular parameters. Although this study is far from the demonstration of a toxic effect of the tested bioactives when administered to humans, it represents a starting point for future research aimed at verifying the existence of a potential hazard due to the wide use of high doses of multiple bioactives.
Collapse
|
14
|
Perumal V, Venkatesan A. Antimicrobial, cytotoxic effect and purification of bacteriocin from vancomycin susceptible Enterococcus faecalis and its safety evaluation for probiotization. Lebensm Wiss Technol 2017. [DOI: 10.1016/j.lwt.2016.12.048] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
15
|
Wang Y, DiSalvo M, Gunasekara DB, Dutton J, Proctor A, Lebhar MS, Williamson IA, Speer J, Howard RL, Smiddy NM, Bultman SJ, Sims CE, Magness ST, Allbritton NL. Self-renewing Monolayer of Primary Colonic or Rectal Epithelial Cells. Cell Mol Gastroenterol Hepatol 2017; 4:165-182.e7. [PMID: 29204504 PMCID: PMC5710741 DOI: 10.1016/j.jcmgh.2017.02.011] [Citation(s) in RCA: 161] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 02/15/2017] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS Three-dimensional organoid culture has fundamentally changed the in vitro study of intestinal biology enabling novel assays; however, its use is limited because of an inaccessible luminal compartment and challenges to data gathering in a three-dimensional hydrogel matrix. Long-lived, self-renewing 2-dimensional (2-D) tissue cultured from primary colon cells has not been accomplished. METHODS The surface matrix and chemical factors that sustain 2-D mouse colonic and human rectal epithelial cell monolayers with cell repertoires comparable to that in vivo were identified. RESULTS The monolayers formed organoids or colonoids when placed in standard Matrigel culture. As with the colonoids, the monolayers exhibited compartmentalization of proliferative and differentiated cells, with proliferative cells located near the peripheral edges of growing monolayers and differentiated cells predominated in the central regions. Screening of 77 dietary compounds and metabolites revealed altered proliferation or differentiation of the murine colonic epithelium. When exposed to a subset of the compound library, murine organoids exhibited similar responses to that of the monolayer but with differences that were likely attributable to the inaccessible organoid lumen. The response of the human primary epithelium to a compound subset was distinct from that of both the murine primary epithelium and human tumor cells. CONCLUSIONS This study demonstrates that a self-renewing 2-D murine and human monolayer derived from primary cells can serve as a physiologically relevant assay system for study of stem cell renewal and differentiation and for compound screening. The platform holds transformative potential for personalized and precision medicine and can be applied to emerging areas of disease modeling and microbiome studies.
Collapse
Key Words
- 2-D, two-dimensional
- 3-D, three-dimensional
- ALP, alkaline phosphatase
- CAG, cytomegalovirus enhancer plus chicken actin promoter
- CI, confidence interval
- Colonic Epithelial Cells
- Compound Screening
- ECM, extracellular matrix
- EDU, 5-ethynyl-2′-deoxyuridine
- EGF, epidermal growth factor
- ENR-W, cell medium with [Wnt-3A] of 30 ng/mL
- ENR-w, cell medium with [Wnt-3A] of 10 ng/mL
- HISC, human intestinal stem cell medium
- IACUC, Institutional Animal Care and Use Committee
- ISC, intestinal stem cell
- Monolayer
- Organoids
- PBS, phosphate-buffered saline
- PDMS, polydimethylsiloxane
- RFP, red fluorescent protein
- SEM, scanning electron microscope
- SSMD, strictly standardized mean difference
- UNC, University of North Carolina
- α-ChgA, anti-chromogranin A
- α-Muc2, anti-mucin2
Collapse
Affiliation(s)
- Yuli Wang
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina
| | - Matthew DiSalvo
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill, and North Carolina State University, Raleigh, North Carolina
| | - Dulan B. Gunasekara
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina
| | - Johanna Dutton
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill, and North Carolina State University, Raleigh, North Carolina
| | - Angela Proctor
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina
| | - Michael S. Lebhar
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill, and North Carolina State University, Raleigh, North Carolina
| | - Ian A. Williamson
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill, and North Carolina State University, Raleigh, North Carolina
| | - Jennifer Speer
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina
| | - Riley L. Howard
- Department of Applied Physical Sciences, University of North Carolina, Chapel Hill, North Carolina
| | - Nicole M. Smiddy
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina
| | - Scott J. Bultman
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina
| | - Christopher E. Sims
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina
| | - Scott T. Magness
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill, and North Carolina State University, Raleigh, North Carolina
| | - Nancy L. Allbritton
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina,Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill, and North Carolina State University, Raleigh, North Carolina,Department of Applied Physical Sciences, University of North Carolina, Chapel Hill, North Carolina,Correspondence Address correspondence to: Nancy L. Allbritton, MD, PhD, Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina 27599. fax: (919) 962-2388.Department of ChemistryUniversity of North CarolinaChapel HillNorth Carolina 27599
| |
Collapse
|
16
|
Devi Avaiyarasi N, David Ravindran A, Venkatesh P, Arul V. In vitro selection, characterization and cytotoxic effect of bacteriocin of Lactobacillus sakei GM3 isolated from goat milk. Food Control 2016. [DOI: 10.1016/j.foodcont.2016.04.036] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
17
|
Characterization of chemical, biological, and antiproliferative properties of fermented black carrot juice, shalgam. Eur Food Res Technol 2016. [DOI: 10.1007/s00217-016-2639-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
18
|
Verbeke KA, Boobis AR, Chiodini A, Edwards CA, Franck A, Kleerebezem M, Nauta A, Raes J, van Tol EAF, Tuohy KM, on behalf of the ILSI Europe Prebiotics Task Force Expert Group ‘Microbial metabolism and fermentation’. Towards microbial fermentation metabolites as markers for health benefits of prebiotics. Nutr Res Rev 2015; 28:42-66. [PMID: 26156216 PMCID: PMC4501371 DOI: 10.1017/s0954422415000037] [Citation(s) in RCA: 225] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Available evidence on the bioactive, nutritional and putative detrimental properties of gut microbial metabolites has been evaluated to support a more integrated view of how prebiotics might affect host health throughout life. The present literature inventory targeted evidence for the physiological and nutritional effects of metabolites, for example, SCFA, the potential toxicity of other metabolites and attempted to determine normal concentration ranges. Furthermore, the biological relevance of more holistic approaches like faecal water toxicity assays and metabolomics and the limitations of faecal measurements were addressed. Existing literature indicates that protein fermentation metabolites (phenol, p-cresol, indole, ammonia), typically considered as potentially harmful, occur at concentration ranges in the colon such that no toxic effects are expected either locally or following systemic absorption. The endproducts of saccharolytic fermentation, SCFA, may have effects on colonic health, host physiology, immunity, lipid and protein metabolism and appetite control. However, measuring SCFA concentrations in faeces is insufficient to assess the dynamic processes of their nutrikinetics. Existing literature on the usefulness of faecal water toxicity measures as indicators of cancer risk seems limited. In conclusion, at present there is insufficient evidence to use changes in faecal bacterial metabolite concentrations as markers of prebiotic effectiveness. Integration of results from metabolomics and metagenomics holds promise for understanding the health implications of prebiotic microbiome modulation but adequate tools for data integration and interpretation are currently lacking. Similarly, studies measuring metabolite fluxes in different body compartments to provide a more accurate picture of their nutrikinetics are needed.
Collapse
Affiliation(s)
- Kristin A. Verbeke
- Translational Research in Gastrointestinal Disorders (TARGID), KU Leuven and Leuven Food Science and Nutrition Research Center (LFoRCe), Leuven, Belgium
| | - Alan R. Boobis
- Department of Medicine, Imperial College London, London, UK
| | - Alessandro Chiodini
- Formerly ILSI Europe, Box 6, Avenue Emmanuel Mounier 83, BE-1200, Brussels, Belgium; now European Commission, Research Executive Agency (REA) Unit B2, Brussels, Belgium
| | - Christine A. Edwards
- Human Nutrition School of Medicine, College of MVLS, University of Glasgow, Glasgow, Scotland
| | | | - Michiel Kleerebezem
- Host Microbe Interactomics, Wageningen University, Wageningen, The Netherlands
| | - Arjen Nauta
- FrieslandCampina, Amersfoort, The Netherlands
| | - Jeroen Raes
- Microbiology and Immunology, Rega Institute, KU Leuven, Leuven; VIB, Leuven; DBIT, Vrije Universiteit Brussel, Brussels, Belgium
| | | | - Kieran M. Tuohy
- Nutrition and Nutrigenomics, Research and Innovation Centre-Fondazione Edmund Mach, Trento, Italy
| | | |
Collapse
|
19
|
Huang XZ, Li ZR, Zhu LB, Huang HY, Hou LL, Lin J. Inhibition of p38 mitogen-activated protein kinase attenuates butyrate-induced intestinal barrier impairment in a Caco-2 cell monolayer model. J Pediatr Gastroenterol Nutr 2014; 59:264-269. [PMID: 24625969 DOI: 10.1097/mpg.0000000000000369] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
OBJECTIVES Butyrate is well known to induce apoptosis in differentiating intestinal epithelial cells. The present study was designed to examine the role of p38 mitogen-activated protein kinase (MAPK) in butyrate-induced intestinal barrier impairment. METHODS The intestinal barrier was determined by measuring the transepithelial electrical resistance (TER) in a Caco-2 cell monolayer model. The permeability was determined by measuring transepithelial passage of fluorescein isothiocyanate-conjugated inulin (inulin-FITC). The morphology of the monolayers was examined with scanning electron microscopy. The apoptosis status was determined by annexin V-FITC labeling and flow cytometry. The activity of p38 MAPK was determined by the phosphorylation status of p38 with Western blotting. RESULTS Butyrate at 5 mM increases the apoptosis rate of Caco-2 cells and induces impairment of intestinal barrier functions as determined by decreased TER and increased inulin-FITC permeability. Butyrate treatment activates p38 MAPK in a concentration- and time-dependent manner. SB203580, a specific p38 inhibitor, inhibits butyrate-induced Caco-2 cell apoptosis. Treatment of SB203580 significantly attenuates the butyrate-induced impairment of barrier functions in the Caco-2 cell monolayer model. CONCLUSIONS p38 MAPK can be activated by butyrate and is involved in the butyrate-induced apoptosis and impairment of intestinal barrier function. Inhibition of p38 MAPK can significantly attenuate butyrate-induced intestinal barrier dysfunction.
Collapse
Affiliation(s)
- Xiao-Zhong Huang
- *Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China †Department of Critical Care Medicine, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China ‡Kravis Children's Hospital of the Icahn School of Medicine at Mount Sinai, New York, NY
| | | | | | | | | | | |
Collapse
|
20
|
Windey K, De Preter V, Louat T, Schuit F, Herman J, Vansant G, Verbeke K. Modulation of protein fermentation does not affect fecal water toxicity: a randomized cross-over study in healthy subjects. PLoS One 2012; 7:e52387. [PMID: 23285019 PMCID: PMC3527498 DOI: 10.1371/journal.pone.0052387] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2012] [Accepted: 11/12/2012] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVE Protein fermentation results in production of metabolites such as ammonia, amines and indolic, phenolic and sulfur-containing compounds. In vitro studies suggest that these metabolites might be toxic. However, human and animal studies do not consistently support these findings. We modified protein fermentation in healthy subjects to assess the effects on colonic metabolism and parameters of gut health, and to identify metabolites associated with toxicity. DESIGN After a 2-week run-in period with normal protein intake (NP), 20 healthy subjects followed an isocaloric high protein (HP) and low protein (LP) diet for 2 weeks in a cross-over design. Protein fermentation was estimated from urinary p-cresol excretion. Fecal metabolite profiles were analyzed using GC-MS and compared using cluster analysis. DGGE was used to analyze microbiota composition. Fecal water genotoxicity and cytotoxicity were determined using the Comet assay and the WST-1-assay, respectively, and were related to the metabolite profiles. RESULTS Dietary protein intake was significantly higher during the HP diet compared to the NP and LP diet. Urinary p-cresol excretion correlated positively with protein intake. Fecal water cytotoxicity correlated negatively with protein fermentation, while fecal water genotoxicity was not correlated with protein fermentation. Heptanal, 3-methyl-2-butanone, dimethyl disulfide and 2-propenyl ester of acetic acid are associated with genotoxicity and indole, 1-octanol, heptanal, 2,4-dithiapentane, allyl-isothiocyanate, 1-methyl-4-(1-methylethenyl)-benzene, propionic acid, octanoic acid, nonanoic acid and decanoic acid with cytotoxicity. CONCLUSION This study does not support a role of protein fermentation in gut toxicity. The identified metabolites can provide new insight into colonic health. TRIAL REGISTRATION ClinicalTrial.gov NCT01280513.
Collapse
Affiliation(s)
- Karen Windey
- Translational Research Center for Gastrointestinal Disorders and Leuven Food Science and Nutrition Research Centre, University Hospital Gasthuisberg, KU Leven Leuven, Belgium
| | - Vicky De Preter
- Translational Research Center for Gastrointestinal Disorders and Leuven Food Science and Nutrition Research Centre, University Hospital Gasthuisberg, KU Leven Leuven, Belgium
| | - Thierry Louat
- Interface Valorisation Platform, KU Leven Leuven, Belgium
| | - Frans Schuit
- Gene Expression Unit, Department of Molecular Cell Biology, KU Leven, Leuven, Belgium
| | - Jean Herman
- Interface Valorisation Platform, KU Leven Leuven, Belgium
| | - Greet Vansant
- Department of Nutrition–Public Health Medicine, Leuven Food Science and Nutrition Research Centre, KU Leven Leuven, Belgium
| | - Kristin Verbeke
- Translational Research Center for Gastrointestinal Disorders and Leuven Food Science and Nutrition Research Centre, University Hospital Gasthuisberg, KU Leven Leuven, Belgium
| |
Collapse
|
21
|
P-glycoprotein induction by breast milk attenuates intestinal inflammation in experimental necrotizing enterocolitis. J Transl Med 2011; 91:1668-79. [PMID: 21788941 PMCID: PMC3909679 DOI: 10.1038/labinvest.2011.113] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
P-glycoprotein (Pgp), a product of the multi-drug resistance gene MDR1a, is a broad specificity efflux ATP cassette transmembrane transporter that is predominantly expressed in epithelial tissues. Because mdr1a(-/-) mice tend to develop spontaneous colitis in bacteria-dependent manner, Pgp is believed to have a role in protection of the intestinal epithelium from luminal bacteria. Here we demonstrate that levels of Pgp in the small intestine of newborn rodents dramatically increase during breastfeeding, but not during formula feeding (FF). In rats and mice, levels of intestinal Pgp peak on days 3-7 and 1-5 of breastfeeding, respectively. The mdr1a(-/-) neonatal mice subjected to FF, hypoxia, and hypothermia have significantly higher incidence and pathology, as well as significantly earlier onset of necrotizing enterocolitis (NEC) than congenic wild type mice. Breast-fed mdr1a(-/-) neonatal mice are also more susceptible to intestinal damage caused by the opportunistic pathogen Cronobacter sakazakii that has been associated with hospital outbreaks of NEC. Breast milk, but not formula, induces Pgp expression in enterocyte cell lines in a dose- and time-dependent manner. High levels of ectopically expressed Pgp protect epithelial cells in vitro from apoptosis induced by C. sakazakii. Taken together, these results show that breast milk-induced expression of Pgp may have a role in the protection of the neonatal intestinal epithelium from injury associated with nascent bacterial colonization.
Collapse
|
22
|
Villarante KI, Elegado FB, Iwatani S, Zendo T, Sonomoto K, de Guzman EE. Purification, characterization and in vitro cytotoxicity of the bacteriocin from Pediococcus acidilactici K2a2-3 against human colon adenocarcinoma (HT29) and human cervical carcinoma (HeLa) cells. World J Microbiol Biotechnol 2010. [DOI: 10.1007/s11274-010-0541-1] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
23
|
Role of calcium signalling and phosphorylations in disruption of the epithelial junctions by Pseudomonas aeruginosa quorum sensing molecule. Eur J Cell Biol 2010; 89:584-97. [DOI: 10.1016/j.ejcb.2010.03.002] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2009] [Revised: 03/09/2010] [Accepted: 03/10/2010] [Indexed: 12/21/2022] Open
|
24
|
Banerjee P, Franz B, Bhunia AK. Mammalian cell-based sensor system. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2010; 117:21-55. [PMID: 20091291 DOI: 10.1007/10_2009_21] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Use of living cells or cellular components in biosensors is receiving increased attention and opens a whole new area of functional diagnostics. The term "mammalian cell-based biosensor" is designated to biosensors utilizing mammalian cells as the biorecognition element. Cell-based assays, such as high-throughput screening (HTS) or cytotoxicity testing, have already emerged as dependable and promising approaches to measure the functionality or toxicity of a compound (in case of HTS); or to probe the presence of pathogenic or toxigenic entities in clinical, environmental, or food samples. External stimuli or changes in cellular microenvironment sometimes perturb the "normal" physiological activities of mammalian cells, thus allowing CBBs to screen, monitor, and measure the analyte-induced changes. The advantage of CBBs is that they can report the presence or absence of active components, such as live pathogens or active toxins. In some cases, mammalian cells or plasma membranes are used as electrical capacitors and cell-cell and cell-substrate contact is measured via conductivity or electrical impedance. In addition, cytopathogenicity or cytotoxicity induced by pathogens or toxins resulting in apoptosis or necrosis could be measured via optical devices using fluorescence or luminescence. This chapter focuses mainly on the type and applications of different mammalian cell-based sensor systems.
Collapse
Affiliation(s)
- Pratik Banerjee
- Laboratory of Food Microbiology & Immunochemistry, Department of Food & Animal Sciences, Alabama A&M University, Normal, AL, 35762, USA
| | | | | |
Collapse
|
25
|
Karlsson C, Ahrné S, Molin G, Berggren A, Palmquist I, Fredrikson GN, Jeppsson B. Probiotic therapy to men with incipient arteriosclerosis initiates increased bacterial diversity in colon: a randomized controlled trial. Atherosclerosis 2009; 208:228-33. [PMID: 19608185 DOI: 10.1016/j.atherosclerosis.2009.06.019] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2009] [Revised: 05/25/2009] [Accepted: 06/09/2009] [Indexed: 12/15/2022]
Abstract
OBJECTIVE This study aimed to clarify the microbial change in the intestinal microbiota in patients, with cardiovascular disease, consuming a drink with high numbers of live Lactobacillus plantarum. METHODS Sixteen males, with atherosclerotic plaque on the carotid wall, were randomly selected from a larger cohort and included in this double blind, placebo controlled study. Colonic biopsies, taken before and after four weeks of probiotic treatment, were analysed with Terminal Restriction Fragment Length Polymorphism, including digestion with MspI and HaeIII. Microbial diversity was calculated, short-chain fatty acids in faeces, and blood markers were analysed. RESULTS Consumption of one probiotic strain of L. plantarum (DSM 9843) increased intestinal microbial diversity. The probiotic group had an increased diversity after consumption of the probiotic drink compared to the change in the placebo group when Shannon and Weaner diversity index (MspI and HaeIII, p=0.026) and Simpson index of diversity (MspI, p=0.044 and HaeIII, p=0.026) were calculated. The fermentation pattern of short-chain fatty acids in faeces were unaffected for most acids, but the probiotic group had decreased concentration of isovaleric acid (p=0.006) and valeric acid (p=0.029). Viable count of lactobacilli increased in the probiotic group (p=0.001), but no significant changes in blood markers were observed. CONCLUSION Administration of a single-strain probiotic increases the bacterial diversity in the gut, and affects the concentration of some short-chain fatty acids. Consumption of the single strain L. plantarum DSM 9843 might be a strategy to favour a diverse intestinal microbiota, which is beneficial for the host.
Collapse
Affiliation(s)
- Caroline Karlsson
- Food Hygiene, Department of Food Technology, Engineering and Nutrition, Lund University, Lund, Sweden
| | | | | | | | | | | | | |
Collapse
|
26
|
Vinolo MAR, Hatanaka E, Lambertucci RH, Newsholme P, Curi R. Effects of short chain fatty acids on effector mechanisms of neutrophils. Cell Biochem Funct 2009; 27:48-55. [PMID: 19107872 DOI: 10.1002/cbf.1533] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Short chain fatty acids (SCFAs) are metabolic by products of anaerobic bacteria fermentation. These fatty acids, despite being an important fuel for colonocytes, are also modulators of leukocyte function. The aim of this study was to evaluate the effects of SCFAs (acetate, propionate, and butyrate) on function of neutrophils, and the possible mechanisms involved. Neutrophils obtained from rats by intraperitoneal lavage 4 h after injection of oyster glycogen solution (1%) were treated with non toxic concentrations of the fatty acids. After that, the following measurements were performed: phagocytosis and destruction of Candida albicans, production of ROS (O(2)(*-), H(2)O(2), and HOCl) and degranulation. Gene expression (p47(phox) and p22(phox)) and protein phosphorylation (p47(phox)) were analyzed by real time reverse transcriptase chain reaction (RT-PCR) and Western blotting, respectively. Butyrate inhibited phagocytosis and killing of C. albicans. This SCFA also had an inhibitory effect on production of O(2)(*-), H(2)O(2), and HOCl by neutrophils stimulated with PMA or fMLP. This effect of butyrate was not caused by modulation of expression of NADPH oxidase subunits (p47(phox) and p22(phox)) but it was in part due to reduced levels of p47(phox) phosphorylation and an increase in the concentration of cyclic AMP. Acetate increased the production of O(2)(*-) and H(2)O(2) in the absence of stimuli but had no effect on phagocytosis and killing of C. albicans. Propionate had no effect on the parameters studied. These results suggest that butyrate can modulate neutrophil function and thus could be important in inflammatory neutrophil-associated diseases.
Collapse
Affiliation(s)
- Marco A R Vinolo
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, São Paulo University, SP, Brazil.
| | | | | | | | | |
Collapse
|
27
|
Vikström E, Bui L, Konradsson P, Magnusson KE. The junctional integrity of epithelial cells is modulated by Pseudomonas aeruginosa quorum sensing molecule through phosphorylation-dependent mechanisms. Exp Cell Res 2008; 315:313-26. [PMID: 19038248 DOI: 10.1016/j.yexcr.2008.10.044] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2008] [Revised: 10/09/2008] [Accepted: 10/30/2008] [Indexed: 01/13/2023]
Abstract
In Pseudomonas aeruginosa, cell-cell communication based on acyl-homoserine lactone (HSL) quorum sensing molecules is known to coordinate the production of virulence factors and biofilms by the bacterium. Incidentally, these bacterial signals can also modulate mammalian cell behaviour. We report that 3O-C(12)-HSL can disrupt adherens junctions in human epithelial Caco-2 cells as evidenced by a reduction of the expression and distribution of E-cadherin and beta-catenin. Using co-immunoprecipitation we also found that P. aeruginosa 3O-C(12)-HSL-treatment resulted in tyrosine hyperphosphorylation of E-cadherin, beta-catenin, occludin and ZO-1. Similarly, serine and threonine residues of E-cadherin and ZO-1 became more phosphorylated after 3O-C(12)-HSL treatment. On the contrary, occludin and beta-catenin underwent dephosphorylation on serine and threonine residues after exposition of 3O-C(12)-HSL. These changes in the phosphorylation state were paralleled by alteration in the structure of junction complexes and increased paracellular permeability. Moreover, pre-treatment of the Caco-2 cells with protein phosphatase and kinase inhibitors prevented 3O-C(12)-HSL-induced changes in paracellular permeability and interactions between occludin-ZO-1 and the E-cadherin-beta-catenin. These findings clearly suggest that an alteration in the phosphorylation status of junction proteins are involved in the changes in cell junction associations and enhanced paracellular permeability, and that bacterial signals are indeed sensed by the host cells.
Collapse
Affiliation(s)
- Elena Vikström
- Division of Medical Microbiology, Department of Clinical and Experimental Medicine, Linköping University, SE-58185 Linköping, Sweden.
| | | | | | | |
Collapse
|
28
|
BHUNIA ARUNK, BANADA PADMAPRIYA, BANERJEE PRATIK, VALADEZ ANGELA, HIRLEMAN EDANIEL. LIGHT SCATTERING, FIBER OPTIC- AND CELL-BASED SENSORS FOR SENSITIVE DETECTION OF FOODBORNE PATHOGENS. ACTA ACUST UNITED AC 2007. [DOI: 10.1111/j.1745-4581.2007.00077.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
29
|
Peng L, He Z, Chen W, Holzman IR, Lin J. Effects of butyrate on intestinal barrier function in a Caco-2 cell monolayer model of intestinal barrier. Pediatr Res 2007; 61:37-41. [PMID: 17211138 DOI: 10.1203/01.pdr.0000250014.92242.f3] [Citation(s) in RCA: 365] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Production of short-chain fatty acids (SCFA) in the intestinal lumen may play an important role in the maintenance of the intestinal barrier. However, overproduction/accumulation of SCFA in the bowel may be toxic to the intestinal mucosa and has been hypothesized to play a role in the pathogenesis of neonatal necrotizing enterocolitis (NEC). By using a Caco-2 cell monolayer model of intestinal barrier, we report here that the effect of butyrate on the intestinal barrier is paradoxical. Butyrate at a low concentration (2 mM) promotes intestinal barrier function as measured by a significant increase in transepithelial electrical resistance (TER) and a significant decrease in inulin permeability. Butyrate at a high concentration (8 mM) reduces TER and increases inulin permeability significantly. Butyrate induces apoptosis and reduces the number of viable Caco-2 cells in a dose-dependent manner. Intestinal barrier function impairment induced by high concentrations of butyrate is most likely related to butyrate-induced cytotoxicity due to apoptosis. We conclude that the effect of butyrate on the intestinal barrier is paradoxical; i.e. whereas low concentrations of butyrate may be beneficial in promoting intestinal barrier function, excessive butyrate may induce severe intestinal epithelial cell apoptosis and disrupt intestinal barrier.
Collapse
Affiliation(s)
- Luying Peng
- Department of Pediatrics, Mount Sinai School of Medicine, New York, New York 10029, USA
| | | | | | | | | |
Collapse
|
30
|
Vikström E, Tafazoli F, Magnusson KE. Pseudomonas aeruginosa quorum sensing molecule N-(3 oxododecanoyl)-l-homoserine lactone disrupts epithelial barrier integrity of Caco-2 cells. FEBS Lett 2006; 580:6921-8. [PMID: 17157842 DOI: 10.1016/j.febslet.2006.11.057] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2006] [Accepted: 11/24/2006] [Indexed: 01/01/2023]
Abstract
Acyl-homoserine lactone (HSL) quorum sensing molecules play an important role in regulation of virulence gene expression in Pseudomonas aeruginosa. Here, we show that 3O-C(12)-HSL can disrupt barrier integrity in human epithelial Caco-2 cells as evidenced by decreased transepithelial electrical resistance (TER), increased paracellular flux, reduction in the expression and distribution of ZO-1 and occludin, and reorganization of F-actin. P. aeruginosa 3O-C(12)-HSL activate p38 and p42/44 kinases, and inhibition of these kinases partly prevented 3O-C(12)-HSL-induced changes in TER, paracellular flux and expression of occludin and ZO-1. These findings demonstrate that P. aeruginosa 3O-C(12)-HSL can modulate tight junction integrity of Caco-2 cells.
Collapse
Affiliation(s)
- Elena Vikström
- Division of Medical Microbiology, Department of Molecular and Clinical Medicine, Linköping University, SE-58185 Linköping, Sweden.
| | | | | |
Collapse
|