51
|
Wang Y, Zhang T, Liu R, Chang M, Wei W, Jin Q, Wang X. New perspective toward nutritional support for malnourished cancer patients: Role of lipids. Compr Rev Food Sci Food Saf 2021; 20:1381-1421. [PMID: 33533186 DOI: 10.1111/1541-4337.12706] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 12/01/2020] [Accepted: 12/28/2020] [Indexed: 12/13/2022]
Abstract
To improve the difficulties related to malnutrition, nutritional support has become an essential part of multidisciplinary comprehensive treatment for cancer. Lipids are essential nutrient source for the human body, and nowadays in clinical practices, it has a positive interventional effect on patients suffering from cancer. However, contribution of lipids in nutritional support of cancer patients is still poorly understood. Moreover, the sensory and physicochemical properties of lipids can severely restrict their applications in lipid-rich formula foods. In this review article, for the first time, we have presented a summary of the existing studies which were related to the associations between different lipids and improved malnutrition in cancer patients and discussed possible mechanisms. Subsequently, we discussed the challenges and effective solutions during processing of lipids into formula foods. Further, by considering existing problems in current lipid nutritional support, we proposed a novel method for the treatment of malnutrition, including developing individualized lipid nutrition for different patients depending on the individual's genotype and enterotype. Nonetheless, this review study provides a new direction for future research on nutritional support and the development of lipid-rich formula foods for cancer patients, and probably will help to improve the efficacy of lipids in the treatment of cancer malnutrition.
Collapse
Affiliation(s)
- Yandan Wang
- National Engineering Research Center for Functional Food, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, State Key Lab of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Tao Zhang
- National Engineering Research Center for Functional Food, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, State Key Lab of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Ruijie Liu
- National Engineering Research Center for Functional Food, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, State Key Lab of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Ming Chang
- National Engineering Research Center for Functional Food, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, State Key Lab of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Wei Wei
- National Engineering Research Center for Functional Food, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, State Key Lab of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Qingzhe Jin
- National Engineering Research Center for Functional Food, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, State Key Lab of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Xingguo Wang
- National Engineering Research Center for Functional Food, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, State Key Lab of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, China
| |
Collapse
|
52
|
Luo J, Lin X, Bordiga M, Brennan C, Xu B. Manipulating effects of fruits and vegetables on gut microbiota – a critical review. Int J Food Sci Technol 2021. [DOI: 10.1111/ijfs.14927] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Jing Luo
- Food Science and Technology Program BNU–HKBU United International College Zhuhai China
| | - Xian Lin
- Food Science and Technology Program BNU–HKBU United International College Zhuhai China
- Guangdong Academy of Agricultural Sciences/Key Laboratory of Functional Foods Ministry of Agriculture and Rural Affairs/Guangdong Key Laboratory of Agricultural Products Processing Sericultural & Agri‐Food Research Institute Guangdong China
| | - Matteo Bordiga
- Dipartimento di Scienze del Farmaco Università degli Studi del Piemonte Orientale ‘A. Avogadro’ Novara Italy
| | - Charles Brennan
- Faculty of Agriculture and Life Sciences Lincoln University Christchurch New Zealand
| | - Baojun Xu
- Food Science and Technology Program BNU–HKBU United International College Zhuhai China
| |
Collapse
|
53
|
Abbasi A, Hajipour N, Hasannezhad P, Baghbanzadeh A, Aghebati-Maleki L. Potential in vivo delivery routes of postbiotics. Crit Rev Food Sci Nutr 2020; 62:3345-3369. [PMID: 33356449 DOI: 10.1080/10408398.2020.1865260] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Bioactive micro- and macro-molecules (postbiotics) derived from gut beneficial microbes are among natural chemical compounds with medical significance. Currently, a unique therapeutic strategy has been developed with an emphasis on the small molecular weight biomolecules that are made by the microbiome, which endow the host with several physiological health benefits. A large number of postbiotics have been characterized, which due to their unique pharmacokinetic properties in terms of controllable aspects of the dosage and various delivery routes, could be employed as promising medical tools since they exert both prevention and treatment strategies in the host. Nevertheless, there are still main challenges for the in vivo delivery of postbiotics. Currently, scientific literature confirms that targeted delivery systems based on nanoparticles, due to their appealing properties in terms of high biocompatibility, biodegradability, low toxicity, and significant capability to carry both hydrophobic and hydrophilic postbiotics, can be used as a novel and safe strategy for targeted delivery or/and release of postbiotics in various (oral, intradermal, and intravenous) in vivo models. The in vivo delivery of postbiotics are in their emerging phase and require massive investigation and randomized double-blind clinical trials if they are to be applied extensively as treatment strategies. This manuscript provides an overview of the various postbiotic metabolites derived from the gut beneficial microbes, their potential therapeutic activities, and recent progressions in the drug delivery field, as well as concisely giving an insight on the main in vivo delivery routes of postbiotics.
Collapse
Affiliation(s)
- Amin Abbasi
- Department of Food Science and Technology, Faculty of Nutrition & Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Neda Hajipour
- Department of Food Science and Technology, Faculty of Nutrition & Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Paniz Hasannezhad
- Department of Medical Engineering Science, University College of Rouzbahan, Sari, Iran
| | - Amir Baghbanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | |
Collapse
|
54
|
Jamka M, Kokot M, Kaczmarek N, Bermagambetova S, Nowak JK, Walkowiak J. The Effect of Sodium Butyrate Enemas Compared with Placebo on Disease Activity, Endoscopic Scores, and Histological and Inflammatory Parameters in Inflammatory Bowel Diseases: A Systematic Review of Randomised Controlled Trials. Complement Med Res 2020; 28:344-356. [PMID: 33352566 DOI: 10.1159/000512952] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 11/10/2020] [Indexed: 01/06/2023]
Abstract
INTRODUCTION Administration of butyrate enemas might improve the health status of patients with inflammatory bowel disease (IBD). However, the results seem equivocal. Therefore, this systematic review aimed to assess the effect of sodium butyrate enemas on disease activity index (DAI), endoscopic scores, as well as histological and inflammatory parameters in IBD patients. METHODS The PubMed, Scopus, Web of Science, and Cochrane databases were searched. Randomised controlled trials published in English that assessed the effect of butyrate enemas on DAI, clinical symptoms, inflammatory markers, as well as histological and endoscopic scores in patients with Crohn's disease (CD) and ulcerative colitis (UC) were included in the analysis. RESULTS Eight studies involving 227 UC patients were included in this analysis. Only one study reported significant differences in DAI between groups. Besides, butyrate treatment groups did not differ significantly from controls concerning the effect on endoscopic and histological scores. Moreover, butyrate enemas exerted a significant effect on few inflammatory parameters measured in colonic mucosal biopsies. CONCLUSION The current evidence is limited and does not support the application of butyrate enemas in UC. There are no reliable data regarding the efficacy of butyrate enemas in CD. The systematic review protocol was registered in the PROSPERO database (CRD42020163654).
Collapse
Affiliation(s)
- Małgorzata Jamka
- Department of Pediatric Gastroenterology and Metabolic Diseases, Poznan University of Medical Sciences, Poznan, Poland
| | - Marta Kokot
- Department of Pediatric Gastroenterology and Metabolic Diseases, Poznan University of Medical Sciences, Poznan, Poland
| | - Nina Kaczmarek
- Department of Pediatric Gastroenterology and Metabolic Diseases, Poznan University of Medical Sciences, Poznan, Poland
| | | | - Jan Krzysztof Nowak
- Department of Pediatric Gastroenterology and Metabolic Diseases, Poznan University of Medical Sciences, Poznan, Poland
| | - Jarosław Walkowiak
- Department of Pediatric Gastroenterology and Metabolic Diseases, Poznan University of Medical Sciences, Poznan, Poland,
| |
Collapse
|
55
|
Hartstra AV, Schüppel V, Imangaliyev S, Schrantee A, Prodan A, Collard D, Levin E, Dallinga-Thie G, Ackermans MT, Winkelmeijer M, Havik SR, Metwaly A, Lagkouvardos I, Nier A, Bergheim I, Heikenwalder M, Dunkel A, Nederveen AJ, Liebisch G, Mancano G, Claus SP, Benítez-Páez A, la Fleur SE, Bergman JJ, Gerdes V, Sanz Y, Booij J, Kemper E, Groen AK, Serlie MJ, Haller D, Nieuwdorp M. Infusion of donor feces affects the gut-brain axis in humans with metabolic syndrome. Mol Metab 2020; 42:101076. [PMID: 32916306 PMCID: PMC7536740 DOI: 10.1016/j.molmet.2020.101076] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/31/2020] [Accepted: 09/04/2020] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVE Increasing evidence indicates that intestinal microbiota play a role in diverse metabolic processes via intestinal butyrate production. Human bariatric surgery data suggest that the gut-brain axis is also involved in this process, but the underlying mechanisms remain unknown. METHODS We compared the effect of fecal microbiota transfer (FMT) from post-Roux-en-Y gastric bypass (RYGB) donors vs oral butyrate supplementation on (123I-FP-CIT-determined) brain dopamine transporter (DAT) and serotonin transporter (SERT) binding as well as stable isotope-determined insulin sensitivity at baseline and after 4 weeks in 24 male and female treatment-naïve metabolic syndrome subjects. Plasma metabolites and fecal microbiota were also determined at these time points. RESULTS We observed an increase in brain DAT after donor FMT compared to oral butyrate that reduced this binding. However, no effect on body weight and insulin sensitivity was demonstrated after post-RYGB donor feces transfer in humans with metabolic syndrome. Increases in fecal levels of Bacteroides uniformis were significantly associated with an increase in DAT, whereas increases in Prevotella spp. showed an inverse association. Changes in the plasma metabolites glycine, betaine, methionine, and lysine (associated with the S-adenosylmethionine cycle) were also associated with altered striatal DAT expression. CONCLUSIONS Although more and larger studies are needed, our data suggest a potential gut microbiota-driven modulation of brain dopamine and serotonin transporters in human subjects with obese metabolic syndrome. These data also suggest the presence of a gut-brain axis in humans that can be modulated. NTR REGISTRATION 4488.
Collapse
Affiliation(s)
- Annick V Hartstra
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Valentina Schüppel
- Chair of Nutrition and Immunology, Technical University of Munich, Freising, Germany
| | - Sultan Imangaliyev
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Anouk Schrantee
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Andrei Prodan
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Didier Collard
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Evgeni Levin
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Geesje Dallinga-Thie
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Mariette T Ackermans
- Laboratory of Endocrinology, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Maaike Winkelmeijer
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Stefan R Havik
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Amira Metwaly
- Chair of Nutrition and Immunology, Technical University of Munich, Freising, Germany
| | - Ilias Lagkouvardos
- ZIEL-Institute for Food and Health, Technical University of Munich, Freising, Germany
| | - Anika Nier
- Department of Nutritional Sciences, Molecular Nutritional Science, University of Vienna, Austria
| | - Ina Bergheim
- Department of Nutritional Sciences, Molecular Nutritional Science, University of Vienna, Austria
| | - Mathias Heikenwalder
- German Cancer Research Center (DKFZ), Division of Chronic Inflammation and Cancer, Heidelberg, Germany
| | - Andreas Dunkel
- Leibniz-Institute for Food Systems Biology, Technical University of Munich, Freising, Germany
| | - Aart J Nederveen
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Gerhard Liebisch
- Department of Laboratory Medicine, University of Regensburg, Regensburg, Germany
| | - Giulia Mancano
- Department of Food and Nutritional Sciences, University of Reading, Reading, United Kingdom
| | - Sandrine P Claus
- Department of Food and Nutritional Sciences, University of Reading, Reading, United Kingdom
| | - Alfonso Benítez-Páez
- Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Valencia, Spain
| | - Susanne E la Fleur
- Laboratory of Endocrinology, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Jacques J Bergman
- Department of Gastroenterology, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Victor Gerdes
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Yolanda Sanz
- Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Valencia, Spain
| | - Jan Booij
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Elles Kemper
- Department of Clinical Pharmacy, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Albert K Groen
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Mireille J Serlie
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Dirk Haller
- Chair of Nutrition and Immunology, Technical University of Munich, Freising, Germany; ZIEL-Institute for Food and Health, Technical University of Munich, Freising, Germany
| | - Max Nieuwdorp
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands.
| |
Collapse
|
56
|
Hernández-Granados MJ, Franco-Robles E. Postbiotics in human health: Possible new functional ingredients? Food Res Int 2020; 137:109660. [DOI: 10.1016/j.foodres.2020.109660] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 08/25/2020] [Accepted: 08/31/2020] [Indexed: 02/06/2023]
|
57
|
Facchin S, Vitulo N, Calgaro M, Buda A, Romualdi C, Pohl D, Perini B, Lorenzon G, Marinelli C, D’Incà R, Sturniolo GC, Savarino EV. Microbiota changes induced by microencapsulated sodium butyrate in patients with inflammatory bowel disease. Neurogastroenterol Motil 2020; 32:e13914. [PMID: 32476236 PMCID: PMC7583468 DOI: 10.1111/nmo.13914] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 04/06/2020] [Accepted: 05/11/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Butyrate has shown anti-inflammatory and regenerative properties, providing symptomatic relief when orally supplemented in patients suffering from various colonic diseases. We investigated the effect of a colonic-delivery formulation of butyrate on the fecal microbiota of patients with inflammatory bowel diseases (IBDs). METHODS In this double-blind, placebo-controlled, pilot study, 49 IBD patients (n = 19 Crohn's disease, CD and n = 30 ulcerative colitis, UC) were randomized to oral administration of microencapsulated-sodium-butyrate (BLM) or placebo for 2 months, in addition to conventional therapy. Eighteen healthy volunteers (HVs) were recruited to provide a healthy microbiota model of the local people. Fecal microbiota from stool samples was assessed by 16S sequencing. Clinical disease activity and quality of life (QoL) were evaluated before and after treatment. KEY RESULTS At baseline, HVs showed a different microbiota composition compared with IBD patients. Sodium-butyrate altered the gut microbiota of IBD patients by increasing bacteria able to produce SCFA in UC patients (Lachnospiraceae spp.) and the butyrogenic colonic bacteria in CD patients (Butyricicoccus). In UC patients, QoL was positively affected by treatment. CONCLUSIONS AND INFERENCES Sodium-butyrate supplementation increases the growth of bacteria able to produce SCFA with potentially anti-inflammatory action. The clinical impact of this finding requires further investigation.
Collapse
Affiliation(s)
- Sonia Facchin
- Department of Surgery, Oncology and Gastroenterology (DISCOG)University Hospital of PaduaPaduaItaly
| | - Nicola Vitulo
- Department of BiotechnologyUniversity of VeronaVeronaItaly
| | - Matteo Calgaro
- Department of BiotechnologyUniversity of VeronaVeronaItaly
| | - Andrea Buda
- Department of Surgery, Oncology and Gastroenterology (DISCOG)University Hospital of PaduaPaduaItaly
| | | | - Daniel Pohl
- Department of GastroenterologyUniversity Hospital ZurichZurichSwitzerland
| | - Barbara Perini
- Department of Surgery, Oncology and Gastroenterology (DISCOG)University Hospital of PaduaPaduaItaly
| | - Greta Lorenzon
- Department of Surgery, Oncology and Gastroenterology (DISCOG)University Hospital of PaduaPaduaItaly
| | - Carla Marinelli
- Department of Surgery, Oncology and Gastroenterology (DISCOG)University Hospital of PaduaPaduaItaly
| | - Renata D’Incà
- Department of Surgery, Oncology and Gastroenterology (DISCOG)University Hospital of PaduaPaduaItaly
| | - Giacomo Carlo Sturniolo
- Department of Surgery, Oncology and Gastroenterology (DISCOG)University Hospital of PaduaPaduaItaly
| | | |
Collapse
|
58
|
Butyrate generated by gut microbiota and its therapeutic role in metabolic syndrome. Pharmacol Res 2020; 160:105174. [PMID: 32860943 DOI: 10.1016/j.phrs.2020.105174] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 08/07/2020] [Accepted: 08/22/2020] [Indexed: 02/07/2023]
Abstract
Metabolic syndrome (MetS) and the associated incidence of cardiovascular disease and type 2 diabetes represents a significant contributor to morbidity and mortality worldwide. Butyrate, a short-chain fatty acid produced by the gut microbiome, has long been known to promote growth in farmed animals and more recently has been reported to improve body weight and composition, lipid profile, insulin sensitivity and glycaemia in animal models of MetS. In vitro studies have examined the influence of butyrate on intestinal cells, adipose tissue, skeletal muscle, hepatocytes, pancreatic islets and blood vessels, highlighting genes and pathways that may contribute to its beneficial effects. Butyrate's influences in these cells have been attributed primarily to its epigenetic effects as a histone deacetylase inhibitor, as well as its role as an agonist of free fatty acid receptors, but clear mechanistic evidence is lacking. There is also uncertainty whether results from animal studies can translate to human trials due to butyrate's poor systemic availability and rapid clearance. Hitherto, several small-scale human clinical trials have failed to show significant benefits in MetS patients. Further trials are clearly needed, including with formulations designed to improve butyrate's availability. Regardless, dietary intervention to increase the rate of butyrate production may be a beneficial addition to current treatment. This review outlines the current body of evidence on the suitability of butyrate supplementation for MetS, looking at mechanistic effects on the various components of MetS and highlighting gaps in the knowledge and roadblocks to its use in humans.
Collapse
|
59
|
Magrin GL, Strauss FJ, Benfatti CAM, Maia LC, Gruber R. Effects of Short-Chain Fatty Acids on Human Oral Epithelial Cells and the Potential Impact on Periodontal Disease: A Systematic Review of In Vitro Studies. Int J Mol Sci 2020; 21:ijms21144895. [PMID: 32664466 PMCID: PMC7402343 DOI: 10.3390/ijms21144895] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 07/03/2020] [Accepted: 07/07/2020] [Indexed: 12/22/2022] Open
Abstract
Short-chain fatty acids (SCFA), bacterial metabolites released from dental biofilm, are supposed to target the oral epithelium. There is, however, no consensus on how SCFA affect the oral epithelial cells. The objective of the present study was to systematically review the available in vitro evidence of the impact of SCFA on human oral epithelial cells in the context of periodontal disease. A comprehensive electronic search using five databases along with a grey literature search was performed. In vitro studies that evaluated the effects of SCFA on human oral epithelial cells were eligible for inclusion. Risk of bias was assessed by the University of Bristol's tool for assessing risk of bias in cell culture studies. Certainty in cumulative evidence was evaluated using GRADE criteria (grading of recommendations assessment, development, and evaluation). Of 3591 records identified, 10 were eligible for inclusion. A meta-analysis was not possible due to the heterogeneity between the studies. The risk of bias across the studies was considered "serious" due to the presence of methodological biases. Despite these limitations, this review showed that SCFA negatively affect the viability of oral epithelial cells by activating a series of cellular events that includes apoptosis, autophagy, and pyroptosis. SCFA impair the integrity and presumably the transmigration of leucocytes through the epithelial layer by changing junctional and adhesion protein expression, respectively. SCFA also affect the expression of chemokines and cytokines in oral epithelial cells. Future research needs to identify the underlying signaling cascades and to translate the in vitro findings into preclinical models.
Collapse
Affiliation(s)
- Gabriel Leonardo Magrin
- Department of Oral Biology, Medical University of Vienna, Sensengasse 2a, 1090 Vienna, Austria; (G.L.M.); (F.J.S.)
- Department of Dentistry, Center for Education and Research on Dental Implants, Federal University of Santa Catarina, Campus Reitor João David Ferreira Lima s/n, Florianopolis 88040-900, Brazil;
| | - Franz Josef Strauss
- Department of Oral Biology, Medical University of Vienna, Sensengasse 2a, 1090 Vienna, Austria; (G.L.M.); (F.J.S.)
- Department of Conservative Dentistry, Faculty of Dentistry, University of Chile, Av. Sergio Livingstone 943, Santiago 7500566, Chile
| | - Cesar Augusto Magalhães Benfatti
- Department of Dentistry, Center for Education and Research on Dental Implants, Federal University of Santa Catarina, Campus Reitor João David Ferreira Lima s/n, Florianopolis 88040-900, Brazil;
| | - Lucianne Cople Maia
- Department of Pediatric Dentistry and Orthodontics, Federal University of Rio de Janeiro, Rua Prof. Rodolpho Paulo Rocco 325, Rio de Janeiro 21941-617, Brazil;
| | - Reinhard Gruber
- Department of Oral Biology, Medical University of Vienna, Sensengasse 2a, 1090 Vienna, Austria; (G.L.M.); (F.J.S.)
- Correspondence:
| |
Collapse
|
60
|
Determination of butyric acid dosage based on clinical and experimental studies - a literature review. GASTROENTEROLOGY REVIEW 2020; 15:119-125. [PMID: 32550943 PMCID: PMC7294979 DOI: 10.5114/pg.2020.95556] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 05/15/2020] [Indexed: 12/12/2022]
Abstract
Short-chain fatty acids produced by bacteria living in the large intestine are the main energy substrate for the colonocytes. Butyric acid is used for the treatment and prevention of exacerbations of various gastrointestinal diseases: diarrhoea, intestinal inflammations, functional disorders, dysbiosis, and post-surgery or post-chemotherapy conditions. The current standard doses of butyric acid (150–300 mg) range between 1.5–3% and 15–30% of the reported daily demand. Increased metabolism of the colonocytes in conditions involving intestine damage or inflammation, increased energy expenditure during a disease, stimulation of intestine growth in ‘stress’ conditions with accelerated intestinal passage and increased intestinal excretion, and decreased production of endogenous butyrate due to changes in bacterial flora in different pathological conditions require a significant increase of the supply of this acid. Physiological high demand for butyrate and known mechanisms of pathological conditions indicate that current supplementation doses do not cover the demand and their increase should be considered.
Collapse
|
61
|
Elnozahi NA, Said EA, Bistawroos AE, Aly RG. Effect of sodium butyrate on gastric ulcer aggravation and hepatic injury inflicted by bile duct ligation in rats. Saudi Pharm J 2020; 28:675-682. [PMID: 32550798 PMCID: PMC7292876 DOI: 10.1016/j.jsps.2020.04.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 04/17/2020] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND AND AIM Cholestasis is positively associated with an increased risk of peptic ulceration. The present study investigated the aggravating effect of cholestasis on piroxicam-induced gastric ulceration. The study also evaluated the effect of sodium butyrate (SoB) on piroxicam-induced gastric ulceration in cholestatic animals and its effect on hepatic tissues and both effects were compared to ursodeoxycholic acid (UDCA) as a standard anticholestatic drug. METHODS Bile duct ligation was adopted for induction of cholestasis in rats. The cholestatic animals received saline, SoB (P.O, 400 mg/kg, twice daily) or UDCA (P.O, 30 mg/kg/day) for 4 days starting from the first day of surgery. On the 4th day, blood samples were collected for determination of serum hepatic markers, then gastric ulcers were induced by piroxicam administration (P.O, 50 mg/kg) and 4 h later, the stomach was isolated and gastric mucosa was collected for biochemical determinations. The ulcer indices for the investigated drugs were compared to omeprazole as a standard acid suppressive drug. RESULTS Piroxicam-induced ulceration was exacerbated in cholestatic rats. Gastric mucosa showed a significant elevation of MDA and TNF-α together with a significant decrease in GSH &VEGF levels. SoB treatment significantly attenuated ulcer development. The afforded protection was higher than that provided by UDCA and was not significantly different from that afforded by omeprazole. SoB significantly decreased gastric mucosal MDA and TNF-α level, whereas UDCA failed to alter these parameters. Both drugs significantly elevated GSH, VEGF and IL10 levels. Similar to UDCA, SoB showed a significant reduction in AST, ALT, GGT, ALP and bilirubin level. Histopathological examination confirmed the attenuating effect of SoB on gastric and hepatic injury. CONCLUSIONS Sodium butyrate effectively protected gastric and hepatic tissues against cholestasis-induced damage. Gastroprotection was mediated through antioxidant, anti-inflammatory and angiogenic activities.
Collapse
Affiliation(s)
- Neveen A. Elnozahi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Egypt
| | - Esraa A. Said
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Egypt
| | - Azza E. Bistawroos
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Egypt
| | - Rania G. Aly
- Department of Pathology, Faculty of Medicine, Alexandria University, Egypt
| |
Collapse
|
62
|
Lamas B, Martins Breyner N, Houdeau E. Impacts of foodborne inorganic nanoparticles on the gut microbiota-immune axis: potential consequences for host health. Part Fibre Toxicol 2020; 17:19. [PMID: 32487227 PMCID: PMC7268708 DOI: 10.1186/s12989-020-00349-z] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 05/11/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND In food toxicology, there is growing interest in studying the impacts of foodborne nanoparticles (NPs, originating from food additives, food supplements or food packaging) on the intestinal microbiome due to the important and complex physiological roles of these microbial communities in host health. Biocidal activities, as described over recent years for most inorganic and metal NPs, could favour chronic changes in the composition and/or metabolic activities of commensal bacteria (namely, intestinal dysbiosis) with consequences on immune functions. Reciprocally, direct interactions of NPs with the immune system (e.g., inflammatory responses, adjuvant or immunosuppressive properties) may in turn have effects on the gut microbiota. Many chronic diseases in humans are associated with alterations along the microbiota-immune system axis, such as inflammatory bowel diseases (IBD) (Crohn's disease and ulcerative colitis), metabolic disorders (e.g., obesity) or colorectal cancer (CRC). This raises the question of whether chronic dietary exposure to inorganic NPs may be viewed as a risk factor facilitating disease onset and/or progression. Deciphering the variety of effects along the microbiota-immune axis may aid the understanding of how daily exposure to inorganic NPs through various foodstuffs may potentially disturb the intricate dialogue between gut commensals and immunity, hence increasing the vulnerability of the host. In animal studies, dose levels and durations of oral treatment are key factors for mimicking exposure conditions to which humans are or may be exposed through the diet on a daily basis, and are needed for hazard identification and risk assessment of foodborne NPs. This review summarizes relevant studies to support the development of predictive toxicological models that account for the gut microbiota-immune axis. CONCLUSIONS The literature indicates that, in addition to evoking immune dysfunctions in the gut, inorganic NPs exhibit a moderate to extensive impact on intestinal microbiota composition and activity, highlighting a recurrent signature that favours colonization of the intestine by pathobionts at the expense of beneficial bacterial strains, as observed in IBD, CRC and obesity. Considering the long-term exposure via food, the effects of NPs on the gut microbiome should be considered in human health risk assessment, especially when a nanomaterial exhibits antimicrobial properties.
Collapse
Affiliation(s)
- Bruno Lamas
- INRAE Toxalim UMR 1331 (Research Center in Food Toxicology), Team Endocrinology and Toxicology of the Intestinal Barrier, INRAE, Toulouse University, ENVT, INP-Purpan, UPS, 180 Chemin de Tournefeuille, 31027, Toulouse cedex 3, France.
| | - Natalia Martins Breyner
- INRAE Toxalim UMR 1331 (Research Center in Food Toxicology), Team Endocrinology and Toxicology of the Intestinal Barrier, INRAE, Toulouse University, ENVT, INP-Purpan, UPS, 180 Chemin de Tournefeuille, 31027, Toulouse cedex 3, France
| | - Eric Houdeau
- INRAE Toxalim UMR 1331 (Research Center in Food Toxicology), Team Endocrinology and Toxicology of the Intestinal Barrier, INRAE, Toulouse University, ENVT, INP-Purpan, UPS, 180 Chemin de Tournefeuille, 31027, Toulouse cedex 3, France.
| |
Collapse
|
63
|
Bai Y, Mansell TJ. Production and Sensing of Butyrate in a Probiotic Escherichia coli Strain. Int J Mol Sci 2020; 21:ijms21103615. [PMID: 32443851 PMCID: PMC7279287 DOI: 10.3390/ijms21103615] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/18/2020] [Accepted: 05/18/2020] [Indexed: 12/11/2022] Open
Abstract
The short-chain fatty acid butyrate plays critical roles in human gut health, affecting immunomodulation, cell differentiation, and apoptosis, while also serving as the preferred carbon source for colon cells. In this work, we have engineered a model probiotic organism, Escherichia coli Nissle 1917 (EcN, serotype O6:K5:H1), to produce butyrate from genomic loci up to approximately 1 g/L (11 mM). Then, for real-time monitoring of butyrate production in cultures, we developed a high-throughput biosensor that responds to intracellular butyrate concentrations, with green fluorescent protein as the reporter. This work provides a foundation for studies of butyrate for therapeutic applications.
Collapse
|
64
|
Verdugo-Meza A, Ye J, Dadlani H, Ghosh S, Gibson DL. Connecting the Dots Between Inflammatory Bowel Disease and Metabolic Syndrome: A Focus on Gut-Derived Metabolites. Nutrients 2020; 12:E1434. [PMID: 32429195 PMCID: PMC7285036 DOI: 10.3390/nu12051434] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/12/2020] [Accepted: 05/13/2020] [Indexed: 02/06/2023] Open
Abstract
The role of the microbiome in health and disease has gained considerable attention and shed light on the etiology of complex diseases like inflammatory bowel disease (IBD) and metabolic syndrome (MetS). Since the microorganisms inhabiting the gut can confer either protective or harmful signals, understanding the functional network between the gut microbes and the host provides a comprehensive picture of health and disease status. In IBD, disruption of the gut barrier enhances microbe infiltration into the submucosae, which enhances the probability that gut-derived metabolites are translocated from the gut to the liver and pancreas. Considering inflammation and the gut microbiome can trigger intestinal barrier dysfunction, risk factors of metabolic diseases such as insulin resistance may have common roots with IBD. In this review, we focus on the overlap between IBD and MetS, and we explore the role of common metabolites in each disease in an attempt to connect a common origin, the gut microbiome and derived metabolites that affect the gut, liver and pancreas.
Collapse
Affiliation(s)
- Andrea Verdugo-Meza
- Department of Biology, University of British Columbia, Okanagan campus, Kelowna, BC V6T 1Z4, Canada; (A.V.-M.); (J.Y.); (H.D.)
| | - Jiayu Ye
- Department of Biology, University of British Columbia, Okanagan campus, Kelowna, BC V6T 1Z4, Canada; (A.V.-M.); (J.Y.); (H.D.)
| | - Hansika Dadlani
- Department of Biology, University of British Columbia, Okanagan campus, Kelowna, BC V6T 1Z4, Canada; (A.V.-M.); (J.Y.); (H.D.)
| | - Sanjoy Ghosh
- Department of Biology, University of British Columbia, Okanagan campus, Kelowna, BC V6T 1Z4, Canada; (A.V.-M.); (J.Y.); (H.D.)
| | - Deanna L. Gibson
- Department of Biology, University of British Columbia, Okanagan campus, Kelowna, BC V6T 1Z4, Canada; (A.V.-M.); (J.Y.); (H.D.)
- Department of Medicine, University of British Columbia, Okanagan campus, Kelowna, BC V1V 1V7, Canada
| |
Collapse
|
65
|
Interactions of dietary fat with the gut microbiota: Evaluation of mechanisms and metabolic consequences. Clin Nutr 2020; 39:994-1018. [DOI: 10.1016/j.clnu.2019.05.003] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 04/25/2019] [Accepted: 05/01/2019] [Indexed: 12/12/2022]
|
66
|
de Groot PF, Nikolic T, Imangaliyev S, Bekkering S, Duinkerken G, Keij FM, Herrema H, Winkelmeijer M, Kroon J, Levin E, Hutten B, Kemper EM, Simsek S, Levels JHM, van Hoorn FA, Bindraban R, Berkvens A, Dallinga-Thie GM, Davids M, Holleman F, Hoekstra JBL, Stroes ESG, Netea M, van Raalte DH, Roep BO, Nieuwdorp M. Oral butyrate does not affect innate immunity and islet autoimmunity in individuals with longstanding type 1 diabetes: a randomised controlled trial. Diabetologia 2020; 63:597-610. [PMID: 31915895 DOI: 10.1007/s00125-019-05073-8] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 11/06/2019] [Indexed: 02/07/2023]
Abstract
AIMS/HYPOTHESIS The pathophysiology of type 1 diabetes has been linked to altered gut microbiota and more specifically to a shortage of intestinal production of the short-chain fatty acid (SCFA) butyrate, which may play key roles in maintaining intestinal epithelial integrity and in human and gut microbial metabolism. Butyrate supplementation can protect against autoimmune diabetes in mouse models. We thus set out to study the effect of oral butyrate vs placebo on glucose regulation and immune variables in human participants with longstanding type 1 diabetes. METHODS We administered a daily oral dose of 4 g sodium butyrate or placebo for 1 month to 30 individuals with longstanding type 1 diabetes, without comorbidity or medication use, in a randomised (1:1), controlled, double-blind crossover trial, with a washout period of 1 month in between. Participants were randomly allocated to the 'oral sodium butyrate capsules first' or 'oral placebo capsules first' study arm in blocks of five. The clinical investigator received blinded medication from the clinical trial pharmacy. All participants, people doing measurements or examinations, or people assessing the outcomes were blinded to group assignment. The primary outcome was a change in the innate immune phenotype (monocyte subsets and in vitro cytokine production). Secondary outcomes were changes in blood markers of islet autoimmunity (cell counts, lymphocyte stimulation indices and CD8 quantum dot assays), glucose and lipid metabolism, beta cell function (by mixed-meal test), gut microbiota and faecal SCFA. The data was collected at the Amsterdam University Medical Centers. RESULTS All 30 participants were analysed. Faecal butyrate and propionate levels were significantly affected by oral butyrate supplementation and butyrate treatment was safe. However, this modulation of intestinal SCFAs did not result in any significant changes in adaptive or innate immunity, or in any of the other outcome variables. In our discussion, we elaborate on this important discrepancy with previous animal work. CONCLUSIONS/INTERPRETATION Oral butyrate supplementation does not significantly affect innate or adaptive immunity in humans with longstanding type 1 diabetes. TRIAL REGISTRATION Netherlands Trial Register: NL4832 (www.trialregister.nl). DATA AVAILABILITY Raw sequencing data are available in the European Nucleotide Archive repository (https://www.ebi.ac.uk/ena/browse) under study PRJEB30292. FUNDING The study was funded by a Le Ducq consortium grant, a CVON grant, a personal ZONMW-VIDI grant and a Dutch Heart Foundation grant.
Collapse
Affiliation(s)
- Pieter F de Groot
- Department of Internal and Vascular Medicine, Academic Medical Center, Meibergdreef 9, Room D3-316, 1105 AZ, Amsterdam, the Netherlands.
| | - Tatjana Nikolic
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands
| | - Sultan Imangaliyev
- Department of Internal and Vascular Medicine, Academic Medical Center, Meibergdreef 9, Room D3-316, 1105 AZ, Amsterdam, the Netherlands
| | - Siroon Bekkering
- Department of Internal and Vascular Medicine, Academic Medical Center, Meibergdreef 9, Room D3-316, 1105 AZ, Amsterdam, the Netherlands
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Gaby Duinkerken
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands
| | - Fleur M Keij
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands
| | - Hilde Herrema
- Department of Internal and Vascular Medicine, Academic Medical Center, Meibergdreef 9, Room D3-316, 1105 AZ, Amsterdam, the Netherlands
| | - Maaike Winkelmeijer
- Department of Internal and Vascular Medicine, Academic Medical Center, Meibergdreef 9, Room D3-316, 1105 AZ, Amsterdam, the Netherlands
| | - Jeffrey Kroon
- Department of Internal and Vascular Medicine, Academic Medical Center, Meibergdreef 9, Room D3-316, 1105 AZ, Amsterdam, the Netherlands
| | - Evgeni Levin
- Department of Internal and Vascular Medicine, Academic Medical Center, Meibergdreef 9, Room D3-316, 1105 AZ, Amsterdam, the Netherlands
| | - Barbara Hutten
- Department of Epidemiology, Amsterdam University Medical Centers, Academic Medical Centre, Amsterdam, the Netherlands
| | - Elles M Kemper
- Clinical Pharmacy, Amsterdam University Medical Centers, Academic Medical Centre, Amsterdam, the Netherlands
| | - Suat Simsek
- Department of Internal Medicine, Alkmaar Medical Center (MCA), Alkmaar, the Netherlands
| | - Johannes H M Levels
- Department of Internal and Vascular Medicine, Academic Medical Center, Meibergdreef 9, Room D3-316, 1105 AZ, Amsterdam, the Netherlands
| | - Flora A van Hoorn
- Department of Internal and Vascular Medicine, Academic Medical Center, Meibergdreef 9, Room D3-316, 1105 AZ, Amsterdam, the Netherlands
| | - Renuka Bindraban
- Department of Internal and Vascular Medicine, Academic Medical Center, Meibergdreef 9, Room D3-316, 1105 AZ, Amsterdam, the Netherlands
| | - Alicia Berkvens
- Department of Internal and Vascular Medicine, Academic Medical Center, Meibergdreef 9, Room D3-316, 1105 AZ, Amsterdam, the Netherlands
| | - Geesje M Dallinga-Thie
- Department of Internal and Vascular Medicine, Academic Medical Center, Meibergdreef 9, Room D3-316, 1105 AZ, Amsterdam, the Netherlands
| | - Mark Davids
- Department of Internal and Vascular Medicine, Academic Medical Center, Meibergdreef 9, Room D3-316, 1105 AZ, Amsterdam, the Netherlands
| | - Frits Holleman
- Department of Internal and Vascular Medicine, Academic Medical Center, Meibergdreef 9, Room D3-316, 1105 AZ, Amsterdam, the Netherlands
| | - Joost B L Hoekstra
- Department of Internal and Vascular Medicine, Academic Medical Center, Meibergdreef 9, Room D3-316, 1105 AZ, Amsterdam, the Netherlands
| | - Erik S G Stroes
- Department of Internal and Vascular Medicine, Academic Medical Center, Meibergdreef 9, Room D3-316, 1105 AZ, Amsterdam, the Netherlands
| | - Mihai Netea
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
- Department for Genomics & Immunoregulation, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| | - Daniël H van Raalte
- Department of Internal and Vascular Medicine, Academic Medical Center, Meibergdreef 9, Room D3-316, 1105 AZ, Amsterdam, the Netherlands
- Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Centers, VU University Medical Centre, Amsterdam, the Netherlands
| | - Bart O Roep
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands
- Department of Diabetes Immunology, Diabetes & Metabolism Research Institute at the Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Max Nieuwdorp
- Department of Internal and Vascular Medicine, Academic Medical Center, Meibergdreef 9, Room D3-316, 1105 AZ, Amsterdam, the Netherlands
- Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Centers, VU University Medical Centre, Amsterdam, the Netherlands
| |
Collapse
|
67
|
Magrin GL, Di Summa F, Strauss FJ, Panahipour L, Mildner M, Magalhães Benfatti CA, Gruber R. Butyrate Decreases ICAM-1 Expression in Human Oral Squamous Cell Carcinoma Cells. Int J Mol Sci 2020; 21:ijms21051679. [PMID: 32121422 PMCID: PMC7084181 DOI: 10.3390/ijms21051679] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 02/20/2020] [Accepted: 02/27/2020] [Indexed: 12/15/2022] Open
Abstract
Short-chain fatty acids (SCFA) are bacterial metabolites that can be found in periodontal pockets. The expression of adhesion molecules such as intercellular adhesion molecule-1 (ICAM-1) within the epithelium pocket is considered to be a key event for the selective transmigration of leucocytes towards the gingival sulcus. However, the impact of SCFA on ICAM-1 expression by oral epithelial cells remains unclear. We therefore exposed the oral squamous carcinoma cell line HSC-2, primary oral epithelial cells and human gingival fibroblasts to SCFA, namely acetate, propionate and butyrate, and stimulated with known inducers of ICAM-1 such as interleukin-1-beta (IL1β) and tumor necrosis factor-alfa (TNFα). We report here that butyrate but not acetate or propionate significantly suppressed the cytokine-induced ICAM-1 expression in HSC-2 epithelial cells and primary epithelial cells. The G-protein coupled receptor-43 (GPR43/ FFAR2) agonist but not the histone deacetylase inhibitor, trichostatin A, mimicked the butyrate effects. Butyrate also attenuated the nuclear translocation of p65 into the nucleus on HSC-2 cells. The decrease of ICAM-1 was independent of Nrf2/HO-1 signaling and phosphorylation of JNK and p38. Nevertheless, butyrate could not reverse an ongoing cytokine-induced ICAM-1 expression in HSC-2 cells. Overall, these observations suggest that butyrate can attenuate cytokine-induced ICAM-1 expression in cells with epithelial origin.
Collapse
Affiliation(s)
- Gabriel Leonardo Magrin
- Department of Oral Biology, School of Dentistry, Medical University of Vienna, Sensengasse 2a, Vienna 1090, Austria; (G.L.M.); (F.D.S.); (F.-J.S.); (L.P.)
- Center for Education and Research on Dental Implants (CEPID), Department of Dentistry, School of Dentistry, Federal University of Santa Catarina, Campus Reitor João David Ferreira Lima s/n, Florianopolis – SC 88040-900, Brazil;
| | - Francesca Di Summa
- Department of Oral Biology, School of Dentistry, Medical University of Vienna, Sensengasse 2a, Vienna 1090, Austria; (G.L.M.); (F.D.S.); (F.-J.S.); (L.P.)
| | - Franz-Josef Strauss
- Department of Oral Biology, School of Dentistry, Medical University of Vienna, Sensengasse 2a, Vienna 1090, Austria; (G.L.M.); (F.D.S.); (F.-J.S.); (L.P.)
- Department of Conservative Dentistry, School of Dentistry, University of Chile, Av. Sergio Livingstone 943, Santiago 7500566, Chile
- Clinic of Reconstructive Dentistry, University of Zurich, 8032 Zurich, Switzerland
| | - Layla Panahipour
- Department of Oral Biology, School of Dentistry, Medical University of Vienna, Sensengasse 2a, Vienna 1090, Austria; (G.L.M.); (F.D.S.); (F.-J.S.); (L.P.)
| | - Michael Mildner
- Department of Dermatology, Medical University of Vienna, Spitalgasse 23, Vienna 1090, Austria;
| | - Cesar Augusto Magalhães Benfatti
- Center for Education and Research on Dental Implants (CEPID), Department of Dentistry, School of Dentistry, Federal University of Santa Catarina, Campus Reitor João David Ferreira Lima s/n, Florianopolis – SC 88040-900, Brazil;
| | - Reinhard Gruber
- Department of Oral Biology, School of Dentistry, Medical University of Vienna, Sensengasse 2a, Vienna 1090, Austria; (G.L.M.); (F.D.S.); (F.-J.S.); (L.P.)
- Department of Periodontology, University Bern, Hochschulstrasse 4, 3012 Bern, Switzerland
- Correspondence:
| |
Collapse
|
68
|
Skin Cutibacterium acnes Mediates Fermentation to Suppress the Calcium Phosphate-Induced Itching: A Butyric Acid Derivative with Potential for Uremic Pruritus. J Clin Med 2020; 9:jcm9020312. [PMID: 31979095 PMCID: PMC7074307 DOI: 10.3390/jcm9020312] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 01/10/2020] [Accepted: 01/20/2020] [Indexed: 12/12/2022] Open
Abstract
Pruritus and inflammation associated with accumulation of calcium phosphate (CaP) under the skin are common problems among dialysis patients with chronic kidney disease (CKD). The role of skin commensal microbiota in the CaP-induced uremic pruritus remains uncharacterized. Skin Cutibacterium acnes (C. acnes) can solubilize CaP by the production of short-chain fatty acids (SCFAs), such as butyric acid, through glucose fermentation. Like butyric acid, the N-[2-(2-Butyrylamino-ethoxy)-ethyl]-butyramide (BA-NH-NH-BA), a butyric acid derivative, remarkably induced acetylation of histone H3 lysine 9 (AcH3K9) in keratinocytes. Topical application of fermenting C. acnes, butyric acid or BA-NH-NH-BA onto mouse skin effectively ameliorated CaP-induced skin itching, interleukin (IL)-6 up-regulation in keratinocytes, and extracellular signal-regulated kinase (ERK) 1/2 activation in dorsal root ganglia (DRG). Activation of ERK 1/2 by CaP was markedly reduced in IL-6 knockout mice. Genus Cutibacterium was detected in relatively low abundance in itchy skin of patients with CKD. Our results identify a role for the skin fermenting C. acnes in ameliorating CaP-induced activation of IL-6/p-ERK signaling and resulting skin inflammation. Furthermore, we provide evidence for the potential therapeutic efficacy of BA-NH-NH-BA as a postbiotic for the treatment of uremic pruritus.
Collapse
|
69
|
Fei Y, Wang Y, Pang Y, Wang W, Zhu D, Xie M, Lan S, Wang Z. Xylooligosaccharide Modulates Gut Microbiota and Alleviates Colonic Inflammation Caused by High Fat Diet Induced Obesity. Front Physiol 2020; 10:1601. [PMID: 32038285 PMCID: PMC6987399 DOI: 10.3389/fphys.2019.01601] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 12/20/2019] [Indexed: 01/13/2023] Open
Abstract
Obesity leads to colonic inflammation and may increase the risk of colorectal cancer. Xylooligosaccharide (XOS) exhibits strong antioxidant and excellent antibacterial properties, and can be utilized by gut microbes to maintain the ecological balance of the intestinal tract. In this study, we explored how XOS modulates the microbiota and regulates high fat diet (HFD) induced inflammation. We measured the changes in body weight and visceral coefficients in rats fed a high-fat diet. We also measured the expression levels of inflammatory factors in the plasma and colonic tissues of the rats using the enzyme-linked immunosorbent assay and real-time quantitative polymerase chain reaction. We analyzed the composition of fecal microorganisms and short chain fatty acid (SCFA) content using 16S rDNA and GC-MS. We found that XOS significantly counteracted HFD induced weight gain. Moreover, the plasma levels of monocyte chemoattractant protein-1, tumor necrosis factor (TNF-α) and lipopolysaccharide decreased in the XOS-treated rats. XOS treatment decreased TNF-α mRNA expression and increased occludin mRNA expression in the rat colon. We observed a reduction in the overall microbial abundance in the feces of the XOS-treated rats, although the proportion of Bacteroidetes/Firmicutes increased significantly and the number of beneficial bacteria increased in the form of dominant microbes. We found that both SCFA-producing bacteria and SCFA content increased in the gut of the XOS-treated rats. We identified a correlation between the abundance of Prevotella and Paraprevotella and SCFA content. Taken together, we propose that XOS can alleviate colonic inflammation by regulating gut microbial composition and enhancing SCFA content in the gut.
Collapse
Affiliation(s)
- Yanquan Fei
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, China
| | - Yan Wang
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, China
| | - Yilin Pang
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, China
| | - Wenyan Wang
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, China
| | - Dan Zhu
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, China
| | - Meigui Xie
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, China
| | - Shile Lan
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, China
| | - Zheng Wang
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, China
| |
Collapse
|
70
|
Ferrer-Picón E, Dotti I, Corraliza AM, Mayorgas A, Esteller M, Perales JC, Ricart E, Masamunt MC, Carrasco A, Tristán E, Esteve M, Salas A. Intestinal Inflammation Modulates the Epithelial Response to Butyrate in Patients With Inflammatory Bowel Disease. Inflamm Bowel Dis 2020; 26:43-55. [PMID: 31211831 PMCID: PMC6905302 DOI: 10.1093/ibd/izz119] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Butyrate-producing gut bacteria are reduced in patients with active inflammatory bowel disease (IBD), supporting the hypothesis that butyrate supplementation may be beneficial in this setting. Nonetheless, earlier studies suggest that the oxidation of butyrate in IBD patients is altered. We propose that inflammation may decrease epithelial butyrate consumption. METHODS Non-IBD controls and IBD patients were recruited for the study. Stool samples were used for short-chain fatty acid and bacterial butyryl CoA:acetate CoA-transferase quantification. Colonic biopsies and ex vivo differentiated epithelial organoids (d-EpOCs) treated with butyrate and/or tumor necrosis factor alpha (TNFα) were used for analyzing the expression of transporters MCT1 and ABCG2, metabolic enzyme ACADS, and butyrate receptor GPR43, and for butyrate metabolism and consumption assays. RESULTS We observed that lower stool content of butyrate-producing bacteria in active IBD patients did not correlate with decreased butyrate concentrations. Indeed, the intestinal epithelial expression of MCT1, ABCG2, ACADS, and GPR43 was altered in active IBD patients. Nonetheless, d-EpOCs derived from IBD patients showed SLC16A1 (gene encoding for MCT1 protein), ABCG2, ACADS, and GPR43 expression levels comparable to controls. Moreover, IBD- and non-IBD-derived d-EpOCs responded similarly to butyrate, as assessed by transcriptional regulation. TNFα significantly altered SLC16A1, ABCG2, and GPR43 transcription in d-EpOCs, mimicking the expression profile observed in biopsies from active IBD patients and resulting in reduced butyrate consumption. CONCLUSIONS We provide evidence that the response to butyrate is not intrinsically altered in IBD patients. However, TNFα renders the epithelium less responsive to this metabolite, defeating the purpose of butyrate supplementation during active inflammation.
Collapse
Affiliation(s)
- Elena Ferrer-Picón
- Department of Gastroenterology, IDIBAPS, Hospital Clínic, CIBERehd, Barcelona, Spain
| | - Isabella Dotti
- Department of Gastroenterology, IDIBAPS, Hospital Clínic, CIBERehd, Barcelona, Spain
| | - Ana M Corraliza
- Department of Gastroenterology, IDIBAPS, Hospital Clínic, CIBERehd, Barcelona, Spain
| | - Aida Mayorgas
- Department of Gastroenterology, IDIBAPS, Hospital Clínic, CIBERehd, Barcelona, Spain
| | - Miriam Esteller
- Department of Gastroenterology, IDIBAPS, Hospital Clínic, CIBERehd, Barcelona, Spain
| | - José Carlos Perales
- Department of Physiological Sciences, Faculty of Medicine, University of Barcelona, L’Hospitalet del Llobregat, Barcelona, Spain
| | - Elena Ricart
- Department of Gastroenterology, IDIBAPS, Hospital Clínic, CIBERehd, Barcelona, Spain
| | - Maria C Masamunt
- Department of Gastroenterology, IDIBAPS, Hospital Clínic, CIBERehd, Barcelona, Spain
| | - Anna Carrasco
- Department of Gastroenterology, Hospital Universitari Mútua Terrassa, CIBERehd, Barcelona, Spain
| | - Eva Tristán
- Department of Gastroenterology, Hospital Universitari Mútua Terrassa, CIBERehd, Barcelona, Spain
| | - Maria Esteve
- Department of Gastroenterology, Hospital Universitari Mútua Terrassa, CIBERehd, Barcelona, Spain
| | - Azucena Salas
- Department of Gastroenterology, IDIBAPS, Hospital Clínic, CIBERehd, Barcelona, Spain,Address correspondence to: Azucena Salas, Department of Gastroenterology, IDIBAPS, Hospital Clínic, CIBERehd, Rosselló 149-153, 08036, Barcelona, Spain ()
| |
Collapse
|
71
|
Red lentil supplementation reduces the severity of dextran sodium sulfate-induced colitis in C57BL/6 male mice. J Funct Foods 2020. [DOI: 10.1016/j.jff.2019.103625] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
|
72
|
Dankar I, Haddarah A, Sepulcre F, Pujolà M. Assessing Mechanical and Rheological Properties of Potato Puree: Effect of Different Ingredient Combinations and Cooking Methods on the Feasibility of 3D Printing. Foods 2019; 9:E21. [PMID: 31878262 PMCID: PMC7023081 DOI: 10.3390/foods9010021] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 12/11/2019] [Accepted: 12/18/2019] [Indexed: 01/10/2023] Open
Abstract
The effects of agar, alginate, butter, olive oil, and carrots on the mechanical and rheological properties of potato puree prepared by two different cooking methods (microwave heating (MP) and boiling (BP)) were investigated and interpreted in terms of starch microstructural changes. Microscopic observations revealed more aggregated and densely concentrated starch granules in MP samples. This consequently led to a significant increase (p < 0.05) in mechanical and rheological properties up to five times more than BP samples. All samples exhibited shear thinning non-Newtonian behavior. Butter proved its ability to maintain stiff network formation with starch molecules forming lipid-starch complexes characterized by high retention properties and increased stability due to high thixotropic and yield stress values. The pseudo-plasticity combined with high self-supporting ability (high yield stress and mechanical values) comprised by MP samples allows them to better behave during advanced technological processes such as extrusion 3D food printing.
Collapse
Affiliation(s)
- Iman Dankar
- Doctoral School of Science and Technology, Lebanese University, EDST, Hadath 1300, Lebanon; (I.D.); (A.H.)
- Departament d’Enginyeria Agroalimentària i Biotecnologia, Universitat Politècnica de Catalunya, BarcelonaTech, 08860 Castelldefels, Spain;
| | - Amira Haddarah
- Doctoral School of Science and Technology, Lebanese University, EDST, Hadath 1300, Lebanon; (I.D.); (A.H.)
| | - Francesc Sepulcre
- Departament d’Enginyeria Agroalimentària i Biotecnologia, Universitat Politècnica de Catalunya, BarcelonaTech, 08860 Castelldefels, Spain;
| | - Montserrat Pujolà
- Departament d’Enginyeria Agroalimentària i Biotecnologia, Universitat Politècnica de Catalunya, BarcelonaTech, 08860 Castelldefels, Spain;
| |
Collapse
|
73
|
Xiao S, Jiang S, Qian D, Duan J. Modulation of microbially derived short-chain fatty acids on intestinal homeostasis, metabolism, and neuropsychiatric disorder. Appl Microbiol Biotechnol 2019; 104:589-601. [PMID: 31865438 DOI: 10.1007/s00253-019-10312-4] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 12/02/2019] [Accepted: 12/10/2019] [Indexed: 12/16/2022]
Abstract
A diverse range of symbiotic gut bacteria codevelops with the host and is considered a metabolic "organ" that not only facilitates harvesting of nutrients from the dietary components but also produces a class of metabolites. Many metabolites of gut microbes have an important impact on host health. For example, an inventory of metabolic intermediates derived from bacterial protein fermentation may affect host physiology and pathophysiology. Additionally, gut microbiota can convert cholesterol to bile acids and further into secondary bile acids which can conversely modulate microbial community. Moreover, new research identifies that microbes synthesize vitamins for us in the colon. Here, we will review data implicating a major class of bacterial metabolites through breaking down dietary fiber we cannot process, short-chain fatty acids (SCFAs), as crucial executors of alteration of immune mechanisms, regulation of metabolic homeostasis, and neuroprotective effects to combat disease and improve health.
Collapse
Affiliation(s)
- Suwei Xiao
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, People's Republic of China
| | - Shu Jiang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, People's Republic of China.
| | - Dawei Qian
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, People's Republic of China
| | - Jinao Duan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, People's Republic of China.
| |
Collapse
|
74
|
Li W, Gelsinger S, Edwards A, Riehle C, Koch D. Changes in meta-transcriptome of rumen epimural microbial community and liver transcriptome in young calves with feed induced acidosis. Sci Rep 2019; 9:18967. [PMID: 31831817 PMCID: PMC6908691 DOI: 10.1038/s41598-019-54055-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 11/05/2019] [Indexed: 12/11/2022] Open
Abstract
The common management practices of dairy calves leads to increased starch concentration in feed, which subsequently may cause rumen acidosis while on milk and during weaning. Until recently, few attempts were undertaken to understand the health risks of prolonged ruminal acidosis in post weaning calves. Resultantly, the molecular changes in the digestive tracts in post-weaning calves with ruminal acidosis remain largely unexplored. In this study, we investigated the liver transcriptome changes along with its correlation with the rumen microbial rRNA expression changes in young calves using our model of feed induced ruminal acidosis. In this model, new born calves were fed a highly processed, starch-rich diet starting from one week of age through 16 weeks. A total of eight calves were involved in this study. Four of them were fed the acidosis-inducing diet (Treated) and the rest of the four were fed a standard starter diet (Control). Liver and rumen epithelial tissues were collected at necropsy at 17 weeks of age. Transcriptome analyses were carried out in the liver tissues and rRNA meta-transcriptome analysis were done using the rumen epithelial tissues. The correlation analysis was performed by comparing the liver mRNA expression with the rumen epithelial rRNA abundance at genus level. Calves with induced ruminal acidosis had significantly lower ruminal pH in comparison to the control group, in addition to significantly less weight-gain over the course of the experiment. In liver tissues, a total of 428 differentially expressed genes (DEGs) (fold-change, FC ≥ 1.5; adjusted P ≤ 0.1) were identified in treated group in comparison to control. Biological pathways enriched by these DEGs included cellular component organization, indicating the impact of ruminal acidosis on liver development in young calves. Specifically, the up-regulated genes were enriched in acute phase response (P < 0.01), pyruvate metabolic process (P < 0.01) and proton-acceptors (P ≪ 0.001), indicating the liver's response to feed induced acidosis at the transcriptome level. Twelve transferase activity related genes had significant correlation with rumen microbial rRNA expression changes. Among these genes, two up-regulated genes were reported with involvement in lipid metabolism in the liver, implying the direct effect of feed-induced acidosis on both the rumen microbial community and liver metabolism. Our study provides insight into the physiological remodeling in the liver resultant from the prolonged acidosis in post weaning calves, which may facilitate future RNA-seq based diagnosis and precision management of rumen acidosis in dairy calves.
Collapse
Affiliation(s)
- Wenli Li
- The Cell Wall Utilization and Biology Laboratory, US Dairy Forage Research Center, USDA ARS, Madison, WI, 53706, USA.
| | - Sonia Gelsinger
- Department of Dairy Science, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Andrea Edwards
- The Cell Wall Utilization and Biology Laboratory, US Dairy Forage Research Center, USDA ARS, Madison, WI, 53706, USA
| | - Christina Riehle
- Department of Genetics, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Daniel Koch
- Department of Computer Engineering, University of Wisconsin-Madison, Madison, WI, 53706, USA
| |
Collapse
|
75
|
Liu Y, Li T, Alim A, Ren D, Zhao Y, Yang X. Regulatory Effects of Stachyose on Colonic and Hepatic Inflammation, Gut Microbiota Dysbiosis, and Peripheral CD4 + T Cell Distribution Abnormality in High-Fat Diet-Fed Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:11665-11674. [PMID: 31588753 DOI: 10.1021/acs.jafc.9b04731] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
A long-term high-fat diet (HFD) can cause a range of health problems. Gut microbiota plays a decisive role in the development of HFD-associated inflammation, involved in function of T cells. This study was designed to probe the regulative effects of dietary stachyose, a functional oligosaccharide, on HFD-induced weight gain, inflammation, gut microbiota dysbiosis, and T cell abnormality in C57Bl/6 mice. Mice were divided into three groups which received normal chow, HFD and HFD plus stachyose (400 mg/kg), respectively. Results showed that administration of stachyose diminished the HFD-induced upregulation of serum TNF-α level and elevation of peripheral blood leukocyte populations to alleviate the HFD-caused colonic and hepatic inflammation in mice. Analysis of gut microbiota revealed that stachyose improved the intestinal homeostasis of HFD-fed mice by improving the bacterial diversity with the increases in the relative abundances of the Prevotellaceae_NK3B31_group, Parasutterella, Christensenellaceae_R-7_group, and Anaerovorax, as well as the fecal level of butanoic acid, while decreasing the ratio of Firmicutes-to-Bacteroidetes and the abundances of the Lachnospiraceae_NK4A136_group, Desulfovibrio, Anaerotruncus, Mucispirillum, Roseburia, and Odoribacter. Flow cytometric analysis showed that stachyose antagonized the HFD-induced decrease of peripheral CD4+ T cell population in mice. Conclusively, these findings suggest that long-term consumption of stachyose can ameliorate the HFD-associated colonic and hepatic inflammation and its complications by modulating gut microbiota.
Collapse
|
76
|
Kimura I, Ichimura A, Ohue-Kitano R, Igarashi M. Free Fatty Acid Receptors in Health and Disease. Physiol Rev 2019; 100:171-210. [PMID: 31487233 DOI: 10.1152/physrev.00041.2018] [Citation(s) in RCA: 488] [Impact Index Per Article: 97.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Fatty acids are metabolized and synthesized as energy substrates during biological responses. Long- and medium-chain fatty acids derived mainly from dietary triglycerides, and short-chain fatty acids (SCFAs) produced by gut microbial fermentation of the otherwise indigestible dietary fiber, constitute the major sources of free fatty acids (FFAs) in the metabolic network. Recently, increasing evidence indicates that FFAs serve not only as energy sources but also as natural ligands for a group of orphan G protein-coupled receptors (GPCRs) termed free fatty acid receptors (FFARs), essentially intertwining metabolism and immunity in multiple ways, such as via inflammation regulation and secretion of peptide hormones. To date, several FFARs that are activated by the FFAs of various chain lengths have been identified and characterized. In particular, FFAR1 (GPR40) and FFAR4 (GPR120) are activated by long-chain saturated and unsaturated fatty acids, while FFAR3 (GPR41) and FFAR2 (GPR43) are activated by SCFAs, mainly acetate, butyrate, and propionate. In this review, we discuss the recent reports on the key physiological functions of the FFAR-mediated signaling transduction pathways in the regulation of metabolism and immune responses. We also attempt to reveal future research opportunities for developing therapeutics for metabolic and immune disorders.
Collapse
Affiliation(s)
- Ikuo Kimura
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Fuchu-shi, Tokyo, Japan; AMED-CREST, Japan Agency for Medical Research and Development, Chiyoda-ku, Tokyo, Japan; and Department of Biochemistry, Kyoto University Graduate School of Pharmaceutical Science, Sakyo, Kyoto, Japan
| | - Atsuhiko Ichimura
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Fuchu-shi, Tokyo, Japan; AMED-CREST, Japan Agency for Medical Research and Development, Chiyoda-ku, Tokyo, Japan; and Department of Biochemistry, Kyoto University Graduate School of Pharmaceutical Science, Sakyo, Kyoto, Japan
| | - Ryuji Ohue-Kitano
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Fuchu-shi, Tokyo, Japan; AMED-CREST, Japan Agency for Medical Research and Development, Chiyoda-ku, Tokyo, Japan; and Department of Biochemistry, Kyoto University Graduate School of Pharmaceutical Science, Sakyo, Kyoto, Japan
| | - Miki Igarashi
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Fuchu-shi, Tokyo, Japan; AMED-CREST, Japan Agency for Medical Research and Development, Chiyoda-ku, Tokyo, Japan; and Department of Biochemistry, Kyoto University Graduate School of Pharmaceutical Science, Sakyo, Kyoto, Japan
| |
Collapse
|
77
|
Monk JM, Wu W, Lepp D, Wellings HR, Hutchinson AL, Liddle DM, Graf D, Pauls KP, Robinson LE, Power KA. Navy bean supplemented high-fat diet improves intestinal health, epithelial barrier integrity and critical aspects of the obese inflammatory phenotype. J Nutr Biochem 2019; 70:91-104. [DOI: 10.1016/j.jnutbio.2019.04.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 04/03/2019] [Accepted: 04/25/2019] [Indexed: 02/07/2023]
|
78
|
Crohn's Disease: Potential Drugs for Modulation of Autophagy. ACTA ACUST UNITED AC 2019; 55:medicina55060224. [PMID: 31146413 PMCID: PMC6630681 DOI: 10.3390/medicina55060224] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 02/26/2019] [Accepted: 05/23/2019] [Indexed: 12/12/2022]
Abstract
Autophagy is an intracellular process whereby cytoplasmic constituents are degraded within lysosomes. Autophagy functions to eliminate unwanted or damaged materials such as proteins and organelles as their accumulation would be harmful to the cellular system. Autophagy also acts as a defense mechanism against invading pathogens and plays an important role in innate and adaptive immunity. In physiological processes, autophagy is involved in the regulation of tissue development, differentiation and remodeling, which are essential for maintaining cellular homeostasis. Recent studies have demonstrated that autophagy is linked to various diseases and involved in pathophysiological roles, such as adaptation during starvation, anti-aging, antigen presentation, tumor suppression and cell death. The modulation of autophagy has shown greatest promise in Crohn’s disease as most of autophagy drugs involved in these diseases are currently under clinical trials and some has been approved by Food and Drug Administration. This review article discusses autophagy and potential drugs that are currently available for its modulation in Crohn’s disease.
Collapse
|
79
|
Torres J, Ellul P, Langhorst J, Mikocka-Walus A, Barreiro-de Acosta M, Basnayake C, Ding NJS, Gilardi D, Katsanos K, Moser G, Opheim R, Palmela C, Pellino G, Van der Marel S, Vavricka SR. European Crohn's and Colitis Organisation Topical Review on Complementary Medicine and Psychotherapy in Inflammatory Bowel Disease. J Crohns Colitis 2019; 13:673-685e. [PMID: 30820529 DOI: 10.1093/ecco-jcc/jjz051] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 02/26/2019] [Indexed: 12/11/2022]
Abstract
Patients with inflammatory bowel disease [IBD] increasingly use alternative and complementary therapies, for which appropriate evidence is often lacking. It is estimated that up to half of all patients with IBD use various forms of complementary and alternative medicine during some point in their disease course. Considering the frequent use of such therapies, it is crucial that physicians and patients are informed about their efficacy and safety in order to provide guidance and evidence-based advice. Additionally, increasing evidence suggests that some psychotherapies and mind-body interventions may be beneficial in the management of IBD, but their best use remains a matter of research. Herein, we provide a comprehensive review of some of the most commonly used complementary, alternative and psychotherapy interventions in IBD.
Collapse
Affiliation(s)
- Joana Torres
- Department of Gastroenterology, Hospital Beatriz Ângelo, Loures, Portugal
| | - Pierre Ellul
- Department of Medicine, Division of Gastroenterology, Mater Dei Hospital, Msida, Malta
| | - Jost Langhorst
- Department of Internal Medicine and Integrative Gastroenterology, Kliniken Essen-Mitte and Chair for Integrative Medicine and Translational Gastroenterology, Klinikum Bamberg, University Duisburg-Essen, Germany
| | | | - Manuel Barreiro-de Acosta
- Department of Gastroenterology, IBD Unit, University Hospital Santiago De Compostela (CHUS), Santiago De Compostela, Spain
| | - Chamara Basnayake
- Department of Gastroenterology, St. Vincent's Hospital Melbourne, Fitzroy, Melbourne, Australia
| | - Nik John Sheng Ding
- Department of Gastroenterology, St. Vincent's Hospital Melbourne, Fitzroy, Melbourne, Australia
| | - Daniela Gilardi
- IBD Centre, Department of Gastroenterology, Humanitas Clinical and Research Institute, Rozzano, Milan, Italy
| | - Konstantinos Katsanos
- Department of Gastroenterology and Hepatology, Division of Internal Medicine, University and Medical School of Ioannina, Ioannina, Greece
| | - Gabriele Moser
- Department of Internal Medicine III, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna, Austria
| | - Randi Opheim
- Department of Gastroenterology, Oslo University Hospital, and Department of Nursing Science, Institute of Health and Society, University of Oslo, Oslo, Norway
| | - Carolina Palmela
- Department of Gastroenterology, Hospital Beatriz Ângelo, Loures, Portugal
| | - Gianluca Pellino
- Department of Advanced Medical and Surgical Sciences, Università degli Studi della Campania "Luigi Vanvitelli", Naples, Italy
| | - Sander Van der Marel
- Department of Gastroenterology and Internal Medicine, Haaglanden Medisch Centrum, The Hague, The Netherlands
| | | |
Collapse
|
80
|
He C, Wang H, Liao WD, Peng C, Shu X, Zhu X, Zhu ZH. Characteristics of mucosa-associated gut microbiota during treatment in Crohn’s disease. World J Gastroenterol 2019; 25:2204-2216. [PMID: 31143071 PMCID: PMC6526154 DOI: 10.3748/wjg.v25.i18.2204] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Revised: 03/25/2019] [Accepted: 04/10/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The dysbiosis of the gut microbiome is evident in Crohn’s disease (CD) compared with healthy controls (HC), although the alterations from active CD to remission after treatment are unclear.
AIM To characterize the mucosa-associated gut microbiota in patients with CD before and after the induction therapy.
METHODS The basic information was collected from the subjects and the CD activity index (CDAI) was calculated in patients. A 16S rRNA sequencing approach was applied to determine the structures of microbial communities in mucosal samples including the terminal ileal, ascending colon, descending colon and rectum. The composition and function of mucosa-associated gut microbiota were compared between samples from the same cohort of patients before and after treatment. Differential taxa were identified to calculate the microbial dysbiosis index (MDI) and the correlation between MDI and CDAI was analyzed using Pearson correlation test. Predictive functional profiling of microbial communities was obtained with PICRUSt.
RESULTS There were no significant differences in microbial richness among the four anatomical sites in individuals. Compared to active disease, the alpha diversity of CD in remission was increased towards the level of HC compared to the active stage. The principal coordinate analysis revealed that samples of active CD were clearly separated from those in remission, which clustered close to HC. Sixty-five genera were identified as differentially abundant between active and quiescent CD, with a loss of Fusobacterium and a gain of potential beneficial bacteria including Lactobacillus, Akkermansia, Roseburia, Ruminococcus and Lachnospira after the induction of remission. The combination of these taxa into a MDI showed a positive correlation with clinical disease severity and a negative correlation with species richness. The increased capacity for the inferred pathways including Lipopolysaccharide biosynthesis and Lipopolysaccharide biosynthesis proteins in patients before treatment negatively correlated with the abundance of Roseburia, Ruminococcus and Lachnospira.
CONCLUSION The dysbiosis of mucosa-associated microbiota was associated with the disease phenotype and may become a potential diagnostic tool for the recurrence of disease.
Collapse
Affiliation(s)
- Cong He
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Huan Wang
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Wang-Di Liao
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Chao Peng
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Xu Shu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Xuan Zhu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Zhen-Hua Zhu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, China
| |
Collapse
|
81
|
Stols-Gonçalves D, Tristão LS, Henneman P, Nieuwdorp M. Epigenetic Markers and Microbiota/Metabolite-Induced Epigenetic Modifications in the Pathogenesis of Obesity, Metabolic Syndrome, Type 2 Diabetes, and Non-alcoholic Fatty Liver Disease. Curr Diab Rep 2019; 19:31. [PMID: 31044315 PMCID: PMC6494784 DOI: 10.1007/s11892-019-1151-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW The metabolic syndrome is a pathological state in which one of the key components is insulin resistance. A wide spectrum of body compartments is involved in its pathophysiology. Genetic and environmental factors such as diet and physical activity are both related to its etiology. Reversible modulation of gene expression without altering the DNA sequence, known as epigenetic modifications, has been shown to drive this complex metabolic cluster of conditions. Here, we aim to examine some of the recent research of specific epigenetically mediated mechanisms and microbiota-induced epigenetic modifications on the development of adipose tissue and obesity, β-cell dysfunction and diabetes, and hepatocytes and non-alcoholic fatty disease. RECENT FINDINGS DNA methylation patterns and histone modifications have been identified in this context; the integrated analysis of genome, epigenome, and transcriptome is likely to expand our knowledge of epigenetics in health and disease. Epigenetic modifications induced by diet-related microbiota or metabolites possibly contribute to the insulin-resistant state. The identification of epigenetic signatures on diabetes and obesity may give us the possibility of developing new interventions, prevention measures, and follow-up strategies.
Collapse
Affiliation(s)
- Daniela Stols-Gonçalves
- Department of Vascular Medicine, Amsterdam UMC, Location AMC, Meibergdreef 9 (Room A01-112), 1105 AZ Amsterdam, The Netherlands
| | - Luca Schiliró Tristão
- Faculdade de Ciências Médicas de Santos (UNILUS), R. Oswaldo Cruz, 179, Boqueirão, Santos, SP 11025-020 Brazil
| | - Peter Henneman
- Department of Clinical Genetics, Amsterdam UMC, Location AMC, Meibergdreef 9 (Room A01-112), 1105 AZ Amsterdam, The Netherlands
| | - Max Nieuwdorp
- Department of Vascular Medicine, Amsterdam UMC, Location AMC, Meibergdreef 9 (Room A01-112), 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
82
|
Verruck S, Balthazar CF, Rocha RS, Silva R, Esmerino EA, Pimentel TC, Freitas MQ, Silva MC, da Cruz AG, Prudencio ES. Dairy foods and positive impact on the consumer's health. ADVANCES IN FOOD AND NUTRITION RESEARCH 2019; 89:95-164. [PMID: 31351531 DOI: 10.1016/bs.afnr.2019.03.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The objective of the present chapter was to demonstrate the state of the art in the recent advances in nutritional and functional components of dairy products research. In this chapter, the main mechanisms responsible and essential for a better understanding of nutritional and functional values of the components of milk and dairy products are highlighted. It also includes a discussion about the positive impacts of fermented milk, cheese, butter, ice cream, and dairy desserts components on the consumer's health.
Collapse
Affiliation(s)
- Silvani Verruck
- Universidade Federal de Santa Catarina (UFSC), Departamento de Ciência e Tecnologia de Alimentos, Florianópolis, Brazil
| | | | - Ramon Silva Rocha
- Universidade Federal Fluminense (UFF), Faculdade de Veterinária, Niterói, Brazil; Instituto Federal de Educação, Ciência e Tecnologia do Rio de Janeiro (IFRJ), Departamento de Alimentos, Rio de Janeiro, Brazil
| | - Ramon Silva
- Universidade Federal Fluminense (UFF), Faculdade de Veterinária, Niterói, Brazil; Instituto Federal de Educação, Ciência e Tecnologia do Rio de Janeiro (IFRJ), Departamento de Alimentos, Rio de Janeiro, Brazil
| | | | | | | | - Marcia Cristina Silva
- Instituto Federal de Educação, Ciência e Tecnologia do Rio de Janeiro (IFRJ), Departamento de Alimentos, Rio de Janeiro, Brazil
| | - Adriano Gomes da Cruz
- Instituto Federal de Educação, Ciência e Tecnologia do Rio de Janeiro (IFRJ), Departamento de Alimentos, Rio de Janeiro, Brazil.
| | - Elane Schwinden Prudencio
- Universidade Federal de Santa Catarina (UFSC), Departamento de Ciência e Tecnologia de Alimentos, Florianópolis, Brazil
| |
Collapse
|
83
|
Tiele A, Wicaksono A, Kansara J, Arasaradnam RP, Covington JA. Breath Analysis Using eNose and Ion Mobility Technology to Diagnose Inflammatory Bowel Disease-A Pilot Study. BIOSENSORS-BASEL 2019; 9:bios9020055. [PMID: 31013848 PMCID: PMC6627846 DOI: 10.3390/bios9020055] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 03/29/2019] [Accepted: 04/08/2019] [Indexed: 12/12/2022]
Abstract
Early diagnosis of inflammatory bowel disease (IBD), including Crohn's disease (CD) and ulcerative colitis (UC), remains a clinical challenge with current tests being invasive and costly. The analysis of volatile organic compounds (VOCs) in exhaled breath and biomarkers in stool (faecal calprotectin (FCP)) show increasing potential as non-invasive diagnostic tools. The aim of this pilot study is to evaluate the efficacy of breath analysis and determine if FCP can be used as an additional non-invasive parameter to supplement breath results, for the diagnosis of IBD. Thirty-nine subjects were recruited (14 CD, 16 UC, 9 controls). Breath samples were analysed using an in-house built electronic nose (Wolf eNose) and commercial gas chromatograph-ion mobility spectrometer (G.A.S. BreathSpec GC-IMS). Both technologies could consistently separate IBD and controls [AUC ± 95%, sensitivity, specificity], eNose: [0.81, 0.67, 0.89]; GC-IMS: [0.93, 0.87, 0.89]. Furthermore, we could separate CD from UC, eNose: [0.88, 0.71, 0.88]; GC-IMS: [0.71, 0.86, 0.62]. Including FCP did not improve distinction between CD vs UC; eNose: [0.74, 1.00, 0.56], but rather, improved separation of CD vs controls and UC vs controls; eNose: [0.77, 0.55, 1.00] and [0.72, 0.89, 0.67] without FCP, [0.81, 0.73, 0.78] and [0.90, 1.00, 0.78] with FCP, respectively. These results confirm the utility of breath analysis to distinguish between IBD-related diagnostic groups. FCP does not add significant diagnostic value to breath analysis within this study.
Collapse
Affiliation(s)
- Akira Tiele
- School of Engineering, University of Warwick, Coventry CV4 7AL, UK.
| | - Alfian Wicaksono
- School of Engineering, University of Warwick, Coventry CV4 7AL, UK.
| | - Jiten Kansara
- Department of Gastroenterology, University Hospitals Coventry and Warwickshire, Coventry CV2 2DX, UK.
| | - Ramesh P Arasaradnam
- Department of Gastroenterology, University Hospitals Coventry and Warwickshire, Coventry CV2 2DX, UK.
- Applied Biological Sciences, Coventry University, Coventry CV1 5FB, UK.
- Health and Life Sciences, University of Leicester, Leicester LE1 7RH, UK.
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK.
| | | |
Collapse
|
84
|
Effects of oral butyrate supplementation on inflammatory potential of circulating peripheral blood mononuclear cells in healthy and obese males. Sci Rep 2019; 9:775. [PMID: 30692581 PMCID: PMC6349871 DOI: 10.1038/s41598-018-37246-7] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 11/30/2018] [Indexed: 12/19/2022] Open
Abstract
Sodium butyrate is well-known for its immune-modulatory properties. Studies until now only focused on the in vitro effects of butyrate or assessed local effects in the gut upon butyrate administration. In this trial, we studied the systemic anti-inflammatory effects induced by sodium butyrate supplementation in humans. Nine healthy (Lean) and ten obese (metabolic syndrome group, MetSyn) males were given 4 grams sodium butyrate daily for 4 weeks. PBMCs were isolated before and after supplementation for direct stimulation experiments and induction of trained immunity by oxidized low-density lipoprotein (oxLDL), β-glucan, or Bacillus Calmette-Guérin vaccine (BCG). Butyrate supplementation moderately affected some of the cytokine responses in the MetSyn group. In the direct stimulation setup, effects of butyrate supplementation were limited. Interestingly, butyrate supplementation decreased oxLDL-induced trained immunity in the MetSyn group for LPS-induced IL-6 responses and Pam3CSK4-induced TNF-α responses. Induction of trained immunity by β-glucan was decreased by butyrate in the MetSyn group for Pam3CSK4-induced IL-10 production. In this study, while having only limited effects on the direct stimulation of cytokine production, butyrate supplementation significantly affected trained immunity in monocytes of obese individuals with metabolic complications. Therefore, oral butyrate supplementation may be beneficial in reducing the overall inflammatory status of circulating monocytes in patients with metabolic syndrome.
Collapse
|
85
|
The Microbiome in Patients With Inflammatory Diseases. Clin Gastroenterol Hepatol 2019; 17:243-255. [PMID: 30196163 DOI: 10.1016/j.cgh.2018.08.078] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 08/17/2018] [Accepted: 08/24/2018] [Indexed: 02/07/2023]
Abstract
Microbial dysbiosis characterized by alterations in the structure and function of the gut microbiome has long been implicated in the pathogenesis of inflammatory bowel disease (IBD). To date, most human IBD microbiome studies are focused on microbial composition rather than function, however, with the latest technical advancements complemented by the rapidly dropping costs, studies focusing on the functional aspects of microbial dysbiosis are on the rise. Several compelling and complimentary pieces of evidence support the notion that the gut microbiome and their metabolites play an important role in the development of IBD. Data from preclinical studies overwhelmingly support the notion that changes in the gut microbiome causally underlie IBD pathogenesis. Hence, there is considerable interest in modulating the state and function of the gut microbiome to achieve therapeutic benefits. While the causal potential of the gut microbiome remains an active area of current research in the clinical setting, accumulating correlative evidence support the view that microbial dysbiosis parallels increased incidence of IBD. In this review, we intend to provide a brief overview of the current human IBD microbiome findings, describe the cause-effect relationships between the gut microbiome and IBD, and discuss the possibility of using microbiome-based approaches in the diagnosis, therapy, and management of disease. In addition, the potential role of microbiome-based interventions in the treatment of human IBD is also discussed.
Collapse
|
86
|
Wasilewska E, Zlotkowska D, Wroblewska B. Yogurt starter cultures of Streptococcus thermophilus and Lactobacillus bulgaricus ameliorate symptoms and modulate the immune response in a mouse model of dextran sulfate sodium-induced colitis. J Dairy Sci 2018; 102:37-53. [PMID: 30343915 DOI: 10.3168/jds.2018-14520] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 08/29/2018] [Indexed: 12/19/2022]
Abstract
We investigated the yogurt starter cultures of Lactobacillus bulgaricus 151 and Streptococcus thermophilus MK-10 for their effect on the severity of experimental colitis, lymphocyte profile, and regulatory T-cell response. Colitis was induced in BALB/c mice via the administration of 3.5% dextran sulfate sodium salt (DSS) in drinking water for 6 d. Next, the mice were gavaged intragastrically with an active yogurt cultures (YC) mixture (∼5 × 109 cfu/mouse per day) or saline (vehicle) for 8 d. Mice receiving DSS or saline alone served as positive and negative controls, respectively. The length of the colon, disease activity index, histological scores, myeloperoxidase activity, epithelium-associated microbes, short-chain fatty acid profile, total IgA antibody-forming cells, CD3+CD8+, CD3+CD4+, CD3+CD4+CD25+, CD3+CD4+CD25+Foxp3+ T-cell subsets, and cytokine profiles (IL-2, IL-4, IL-6, IL-10, IL-17A, IFN-γ, and tumor necrosis factor) were examined after termination of the mice. Feeding mice with YC mixture reduced disease symptoms and modified intestinal microbiota and host inflammatory responsiveness to DSS. We observed limited weight loss and a decreased disease activity index score, lowered myeloperoxidase activity, and somewhat reduced damage of the intestine. The YC mixture upregulated the colon length, increased the amount and diversity of mucosa-associated microbes (enterobacteria, enterococci, and yeast), and decreased the concentration of putrefactive short-chain fatty acids in the cecal contents. It downregulated the input of cytotoxic CD3+CD8+ T cells and CD3+CD4+CD25+FoxP3+ regulatory T cells in Peyer's patches and enhanced CD3+CD4+CD25+ T cells in spleens and CD3+CD4+CD25+FoxP3+ cells in peripheral blood mononuclear cells. Simultaneously, IgA antibody-forming cells were downregulated in mesenteric lymph nodes (MLN) and enhanced in spleens (SPL). The cultures mostly enhanced the production of cytokines tested in MLN and SPL, except for IL-6, which was downregulated in MLN. Interleukin-2 and IL-4 were the most upregulated in MLN, whereas IL-10, IL-4, IL-2, IFN-γ, and tumor necrosis factor were most upregulated in SPL. In serum, the YC mixture downregulated IFN-γ and clearly increased IL-2. Based on these results, we recognize the high anti-inflammatory and immunomodulatory potential of the L. bulgaricus 151 and S. thermophilus MK-10 set. The strains possess the ability to modulate the intestinal mucosal and systemic immune system toward both IgA production and induction of regulatory T cells, shifting Th1/Th2 balance.
Collapse
Affiliation(s)
- E Wasilewska
- Department of Immunology and Food Microbiology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, 10-748 Olsztyn, Poland.
| | - D Zlotkowska
- Department of Immunology and Food Microbiology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, 10-748 Olsztyn, Poland
| | - B Wroblewska
- Department of Immunology and Food Microbiology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, 10-748 Olsztyn, Poland
| |
Collapse
|
87
|
Monk JM, Wu W, McGillis LH, Wellings HR, Hutchinson AL, Liddle DM, Graf D, Robinson LE, Power KA. Chickpea supplementation prior to colitis onset reduces inflammation in dextran sodium sulfate-treated C57Bl/6 male mice. Appl Physiol Nutr Metab 2018; 43:893-901. [PMID: 29522694 DOI: 10.1139/apnm-2017-0689] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The potential for a chickpea-supplemented diet (rich in fermentable nondigestible carbohydrates and phenolic compounds) to modify the colonic microenvironment and attenuate the severity of acute colonic inflammation was investigated. C57Bl/6 male mice were fed a control basal diet or basal diet supplemented with 20% cooked chickpea flour for 3 weeks prior to acute colitis onset induced by 7-day exposure to dextran sodium sulfate (DSS; 2% w/v in drinking water) and colon and serum levels of inflammatory mediators were assessed. Despite an equal degree of DSS-induced epithelial barrier histological damage and clinical symptoms between dietary groups, biomarkers of the ensuing inflammatory response were attenuated by chickpea pre-feeding, including reduced colon tissue activation of nuclear factor kappa B and inflammatory cytokine production (tumor necrosis factor alpha and interleukin (IL)-18). Additionally, colon protein expression of anti-inflammatory (IL-10) and epithelial repair (IL-22 and IL-27) cytokines were increased by chickpea pre-feeding. Furthermore, during acute colitis, chickpea pre-feeding increased markers of enhanced colonic function, including Relmβ and IgA gene expression. Collectively, chickpea pre-feeding modulated the baseline function of the colonic microenvironment, whereby upon induction of acute colitis, the severity of the inflammatory response was attenuated.
Collapse
Affiliation(s)
- Jennifer M Monk
- a Guelph Research and Development Centre, Agriculture and Agri-Food Canada, Guelph, ON N1G 5C9, Canada
- b Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Wenqing Wu
- a Guelph Research and Development Centre, Agriculture and Agri-Food Canada, Guelph, ON N1G 5C9, Canada
| | - Laurel H McGillis
- b Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Hannah R Wellings
- b Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Amber L Hutchinson
- b Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Danyelle M Liddle
- b Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Daniela Graf
- a Guelph Research and Development Centre, Agriculture and Agri-Food Canada, Guelph, ON N1G 5C9, Canada
- b Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Lindsay E Robinson
- b Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Krista A Power
- a Guelph Research and Development Centre, Agriculture and Agri-Food Canada, Guelph, ON N1G 5C9, Canada
- b Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON N1G 2W1, Canada
| |
Collapse
|
88
|
Westfall S, Lomis N, Prakash S. A polyphenol-rich prebiotic in combination with a novel probiotic formulation alleviates markers of obesity and diabetes in Drosophila. J Funct Foods 2018. [DOI: 10.1016/j.jff.2018.07.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
89
|
Yan HM, Zhao HJ, Guo DY, Zhu PQ, Zhang CL, Jiang W. Gut microbiota alterations in moderate to severe acne vulgaris patients. J Dermatol 2018; 45:1166-1171. [PMID: 30101990 DOI: 10.1111/1346-8138.14586] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 06/20/2018] [Indexed: 01/06/2023]
Abstract
Acne vulgaris is a chronic inflammatory dermatosis affecting approximately 85% of adolescents. There are many factors contributing to the development of this ailment. A recent study indicated that gut microbiota takes part in the pathogenesis of acne. We aimed to investigate the link between acne vulgaris and gut microbiota. A total of 31 moderate to severe acne vulgaris patients and 31 healthy controls were enrolled. We collected their feces, and gut microbiota was evaluated by the hypervariable regions of 16S rRNA genes through high-throughput sequencing. We identified links between acne vulgaris and changes of gut microbiota. At the phylum level, Actinobacteria (0.89% in acne patients and 2.84% in normal controls, P = 0.004) was decreased and Proteobacteria (8.35% in acne patients and 7.01% in normal controls, P = 0.031) was increased. At the genus level, Bifidobacterium, Butyricicoccus, Coprobacillus, Lactobacillus and Allobaculum were all decreased. The observed difference in genera between acne patients and healthy controls provides a new insight into the link between gut microbiota changes and acne vulgaris risk.
Collapse
Affiliation(s)
- Hui-Min Yan
- Department of Dermatology, Peking University Third Hospital, Beijing, China
| | - Hui-Juan Zhao
- Department of Dermatology, Peking University Third Hospital, Beijing, China
| | - Du-Yi Guo
- Department of Dermatology, Peking University Third Hospital, Beijing, China
| | - Pei-Qiu Zhu
- Department of Dermatology, Peking University Third Hospital, Beijing, China
| | - Chun-Lei Zhang
- Department of Dermatology, Peking University Third Hospital, Beijing, China
| | - Wei Jiang
- Department of Dermatology, Peking University Third Hospital, Beijing, China
| |
Collapse
|
90
|
Fang X. Impaired tissue barriers as potential therapeutic targets for Parkinson's disease and amyotrophic lateral sclerosis. Metab Brain Dis 2018; 33:1031-1043. [PMID: 29681010 DOI: 10.1007/s11011-018-0239-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 04/13/2018] [Indexed: 12/12/2022]
Abstract
The blood-brain barrier and the intestinal barrier show signs of disruption in patients with idiopathic Parkinson's disease (PD) and animal models of nigrostriatal degeneration, and likewise in amyotrophic lateral sclerosis (ALS) models. A substantial body of evidence shows that defects in epithelial membrane barriers, both in the gut and within the cerebral vasculature, can result in increased vulnerability of tissues to external factors potentially participating in the pathogenesis of PD and ALS. As such, restoration of tissue barriers may prove to be a novel therapeutic target in neurodegenerative disease. In this review, we focus on the potential of new intervention strategies for rescuing and maintaining barrier functions in PD and ALS.
Collapse
Affiliation(s)
- Xin Fang
- Department of Neurology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China.
| |
Collapse
|
91
|
Bauer E, Thiele I. From metagenomic data to personalized in silico microbiotas: predicting dietary supplements for Crohn's disease. NPJ Syst Biol Appl 2018; 4:27. [PMID: 30083388 PMCID: PMC6068170 DOI: 10.1038/s41540-018-0063-2] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 05/17/2018] [Accepted: 05/30/2018] [Indexed: 12/13/2022] Open
Abstract
Crohn's disease (CD) is associated with an ecological imbalance of the intestinal microbiota, consisting of hundreds of species. The underlying complexity as well as individual differences between patients contributes to the difficulty to define a standardized treatment. Computational modeling can systematically investigate metabolic interactions between gut microbes to unravel mechanistic insights. In this study, we integrated metagenomic data of CD patients and healthy controls with genome-scale metabolic models into personalized in silico microbiotas. We predicted short chain fatty acid (SFCA) levels for patients and controls, which were overall congruent with experimental findings. As an emergent property, low concentrations of SCFA were predicted for CD patients and the SCFA signatures were unique to each patient. Consequently, we suggest personalized dietary treatments that could improve each patient's SCFA levels. The underlying modeling approach could aid clinical practice to find dietary treatment and guide recovery by rationally proposing food aliments.
Collapse
Affiliation(s)
- Eugen Bauer
- Luxembourg Centre for Systems Biomedicine, Universite du Luxembourg, Esch-sur-Alzette, Luxembourg, L-4362 Luxembourg
| | - Ines Thiele
- Luxembourg Centre for Systems Biomedicine, Universite du Luxembourg, Esch-sur-Alzette, Luxembourg, L-4362 Luxembourg
| |
Collapse
|
92
|
Gill PA, van Zelm MC, Muir JG, Gibson PR. Review article: short chain fatty acids as potential therapeutic agents in human gastrointestinal and inflammatory disorders. Aliment Pharmacol Ther 2018; 48:15-34. [PMID: 29722430 DOI: 10.1111/apt.14689] [Citation(s) in RCA: 304] [Impact Index Per Article: 50.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 02/09/2018] [Accepted: 04/06/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND Butyrate, propionate and acetate are short chain fatty acids (SCFA), important for maintaining a healthy colon and are considered as protective in colorectal carcinogenesis. However, they may also regulate immune responses and the composition of the intestinal microbiota. Consequently, their importance in a variety of chronic inflammatory diseases is emerging. AIMS To review the physiology and metabolism of SCFA in humans, cellular and molecular mechanisms by which SCFA may act in health and disease, and approaches for therapeutic delivery of SCFA. METHODS A PubMed literature search was conducted for clinical and pre-clinical studies using search terms: 'dietary fibre', short-chain fatty acids', 'acetate', 'propionate', 'butyrate', 'inflammation', 'immune', 'gastrointestinal', 'metabolism'. RESULTS A wide range of pre-clinical evidence supports roles for SCFA as modulators of not only colonic function, but also multiple inflammatory and metabolic processes. SCFA are implicated in many autoimmune, allergic and metabolic diseases. However, translating effects of SCFA from animal studies to human disease is limited by physiological and dietary differences and by the challenge of delivering sufficient amounts of SCFA to the target sites that include the colon and the systemic circulation. Development of novel targeted approaches for colonic delivery, combined with postbiotic supplementation, may represent desirable strategies to achieve adequate targeted SCFA delivery. CONCLUSIONS There is a large array of potential disease-modulating effects of SCFA. Adequate targeted delivery to the sites of action is the main limitation of such application. The ongoing development and evaluation of novel delivery techniques offer potential for translating promise to therapeutic benefit.
Collapse
Affiliation(s)
- P A Gill
- Department of Gastroenterology, Central Clinical School, Monash University and Alfred Hospital, Melbourne, Vic., Australia.,Department of Immunology and Pathology, Central Clinical School, Monash University and Alfred Hospital, Melbourne, Vic, Australia
| | - M C van Zelm
- Department of Immunology and Pathology, Central Clinical School, Monash University and Alfred Hospital, Melbourne, Vic, Australia
| | - J G Muir
- Department of Gastroenterology, Central Clinical School, Monash University and Alfred Hospital, Melbourne, Vic., Australia
| | - P R Gibson
- Department of Gastroenterology, Central Clinical School, Monash University and Alfred Hospital, Melbourne, Vic., Australia
| |
Collapse
|
93
|
Differential metabolic effects of oral butyrate treatment in lean versus metabolic syndrome subjects. Clin Transl Gastroenterol 2018; 9:155. [PMID: 29799027 PMCID: PMC5968024 DOI: 10.1038/s41424-018-0025-4] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 04/22/2018] [Indexed: 12/13/2022] Open
Abstract
Background Gut microbiota-derived short-chain fatty acids (SCFAs) have been associated with beneficial metabolic effects. However, the direct effect of oral butyrate on metabolic parameters in humans has never been studied. In this first in men pilot study, we thus treated both lean and metabolic syndrome male subjects with oral sodium butyrate and investigated the effect on metabolism. Methods Healthy lean males (n = 9) and metabolic syndrome males (n = 10) were treated with oral 4 g of sodium butyrate daily for 4 weeks. Before and after treatment, insulin sensitivity was determined by a two-step hyperinsulinemic euglycemic clamp using [6,6-2H2]-glucose. Brown adipose tissue (BAT) uptake of glucose was visualized using 18F-FDG PET-CT. Fecal SCFA and bile acid concentrations as well as microbiota composition were determined before and after treatment. Results Oral butyrate had no effect on plasma and fecal butyrate levels after treatment, but did alter other SCFAs in both plasma and feces. Moreover, only in healthy lean subjects a significant improvement was observed in both peripheral (median Rd: from 71 to 82 µmol/kg min, p < 0.05) and hepatic insulin sensitivity (EGP suppression from 75 to 82% p < 0.05). Although BAT activity was significantly higher at baseline in lean (SUVmax: 12.4 ± 1.8) compared with metabolic syndrome subjects (SUVmax: 0.3 ± 0.8, p < 0.01), no significant effect following butyrate treatment on BAT was observed in either group (SUVmax lean to 13.3 ± 2.4 versus metabolic syndrome subjects to 1.2 ± 4.1). Conclusions Oral butyrate treatment beneficially affects glucose metabolism in lean but not metabolic syndrome subjects, presumably due to an altered SCFA handling in insulin-resistant subjects. Although preliminary, these first in men findings argue against oral butyrate supplementation as treatment for glucose regulation in human subjects with type 2 diabetes mellitus.
Collapse
|
94
|
Potential Synergies of β-Hydroxybutyrate and Butyrate on the Modulation of Metabolism, Inflammation, Cognition, and General Health. J Nutr Metab 2018; 2018:7195760. [PMID: 29805804 PMCID: PMC5902005 DOI: 10.1155/2018/7195760] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 02/06/2018] [Accepted: 02/28/2018] [Indexed: 12/31/2022] Open
Abstract
The low-carbohydrate high-fat diet (LCHFD), also known as the ketogenic diet, has cycled in and out of popularity for decades as a therapeutic program to treat metabolic syndrome, weight mismanagement, and drug-resistant disorders as complex as epilepsy, cancer, dementia, and depression. Despite the benefits of this diet, health care professionals still question its safety due to the elevated serum ketones it induces and the limited dietary fiber. To compound the controversy, patient compliance with the program is poor due to the restrictive nature of the diet and symptoms related to energy deficit and gastrointestinal adversity during the introductory and energy substrate transition phase of the diet. The studies presented here demonstrate safety and efficacy of the diet including the scientific support and rationale for the administration of exogenous ketone bodies and ketone sources as a complement to the restrictive dietary protocol or as an alternative to the diet. This review also highlights the synergy provided by exogenous ketone, β-hydroxybutyrate (BHB), accompanied by the short chain fatty acid, butyrate (BA) in the context of cellular and physiological outcomes. More work is needed to unveil the molecular mechanisms by which this program provides health benefits.
Collapse
|
95
|
Navy and black bean supplementation attenuates colitis-associated inflammation and colonic epithelial damage. J Nutr Biochem 2018; 56:215-223. [PMID: 29631142 DOI: 10.1016/j.jnutbio.2018.02.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 01/15/2018] [Accepted: 02/21/2018] [Indexed: 12/15/2022]
Abstract
The enriched levels of nondigestible fermentable carbohydrates and phenolic compounds found in common beans can exert immunomodulatory effects within the colon that improve gut health and mitigate the severity of colitis-associated inflammatory pathology. Prior to acute colitis onset, C57Bl/6 mice were prefed isocaloric 20% cooked navy bean (NB) or black bean (BB) diets for 3 weeks and switched to control basal diet (BD) 24 h prior to colitis induction via 5-day exposure to dextran sodium sulfate (2% w/v in drinking water)+3 days of fresh water. The severity of the acute colitis phenotype was attenuated by bean prefeeding, evidenced by reduced colon tissue inflammatory transcription factor activation (NFκB, STAT3) and inflammatory mediator levels in the colon (IL-1β, IL-6, IL-18 and MCP-1) and serum (TNFα, IL-6, IL-1β, MCP-1) versus BD (P≤.05). Additionally, biomarkers of enhanced wound repair responses were increased by bean prefeeding including colon tissue protein levels of IL-22, IL-27 and activated (i.e., GTP-bound) Cdc42 and Rac1 versus BD (P≤.05). mRNA expressions of genes involved in normal colonic epithelial function and the promotion of epithelial barrier integrity, defense and/or restitution and wound closure including MUC1, RELMβ, IgA and REG3γ were all increased in NB and BB prefed mice versus BD (P≤.05). Collectively, bean supplementation prior to colitis induction (i.e., mimicking disease relapse) primes the colonic microenvironment to attenuate the severity of the colitis inflammatory phenotype and maintain aspects of epithelial barrier function.
Collapse
|
96
|
Shi H, Chang Y, Gao Y, Wang X, Chen X, Wang Y, Xue C, Tang Q. Dietary fucoidan of Acaudina molpadioides alters gut microbiota and mitigates intestinal mucosal injury induced by cyclophosphamide. Food Funct 2018; 8:3383-3393. [PMID: 28861559 DOI: 10.1039/c7fo00932a] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Cyclophosphamide (cy) is a widely used cancer drug. Many researchers have focused on the prevention and alleviation of its side effects, particularly damage to the intestinal mucosal barrier. In this study, we examined the effects of fucoidan, isolated from Acaudina molpadioides, on mice with intestinal mucosal damage induced by cyclophosphamide. Our results showed that fucoidan intervention could relieve injury such as decreasing inflammation and increasing the expression of tight junction proteins, and 50 kDa fucoidan significantly increased the abundance of short chain fatty acid (SCFA) producer Coprococcus, Rikenella, and Butyricicoccus (p < 0.05, p < 0.001, and p < 0.05, respectively) species within the intestinal mucosa compared with the cyclophosphamide group, as determined by 16S rDNA gene high-throughput sequencing. In addition, SCFAs, particularly propionate, butyrate, and total SCFAs, were increased in the feces, and SCFA receptors were upregulated in the small intestine. The protective effects of fucoidan on cyclophosphamide treatment may be associated with gut microflora, and 50 kDa fucoidan had superior effects. Therefore, fucoidan may have applications as an effective supplement to protect against intestinal mucosal barrier damage during chemotherapy.
Collapse
Affiliation(s)
- Hongjie Shi
- College of Food Science and Engineering, Ocean University of China, Qingdao, China.
| | | | | | | | | | | | | | | |
Collapse
|
97
|
Laserna-Mendieta EJ, Clooney AG, Carretero-Gomez JF, Moran C, Sheehan D, Nolan JA, Hill C, Gahan CGM, Joyce SA, Shanahan F, Claesson MJ. Determinants of Reduced Genetic Capacity for Butyrate Synthesis by the Gut Microbiome in Crohn's Disease and Ulcerative Colitis. J Crohns Colitis 2018; 12:204-216. [PMID: 29373727 DOI: 10.1093/ecco-jcc/jjx137] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 10/05/2017] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIMS Alterations in short chain fatty acid metabolism, particularly butyrate, have been reported in inflammatory bowel disease, but results have been conflicting because of small study numbers and failure to distinguish disease type, activity or other variables such as diet. We performed a comparative assessment of the capacity of the microbiota for butyrate synthesis, by quantifying butyryl-CoA:acetate CoA-transferase [BCoAT] gene content in stool from patients with Crohn's disease [CD; n = 71], ulcerative colitis [UC; n = 58] and controls [n = 75], and determined whether it was related to active vs inactive inflammation, microbial diversity, and composition and/or dietary habits. METHODS BCoAT gene content was quantified by quantitative polymerase chain reaction [qPCR]. Disease activity was assessed clinically and faecal calprotectin concentration measured. Microbial composition was determined by sequencing 16S rRNA gene. Dietary data were collected using an established food frequency questionnaire. RESULTS Reduced butyrate-synthetic capacity was found in patients with active and inactive CD [p < 0.001 and p < 0.01, respectively], but only in active UC [p < 0.05]. In CD, low BCoAT gene content was associated with ileal location, stenotic behaviour, increased inflammation, lower microbial diversity, greater microbiota compositional change, and decreased butyrogenic taxa. Reduced BCoAT gene content in patients with CD was linked with a different regimen characterised by lower dietary fibre. CONCLUSIONS Reduced butyrate-synthetic capacity of the microbiota is more evident in CD than UC and may relate to reduced fibre intake. The results suggest that simple replacement of butyrate per se may be therapeutically inadequate, whereas manipulation of microbial synthesis, perhaps by dietary means, may be more appropriate.
Collapse
Affiliation(s)
- Emilio J Laserna-Mendieta
- APC Microbiome Institute, University College Cork, Cork, Ireland.,School of Microbiology, University College Cork, Cork, Ireland
| | - Adam G Clooney
- APC Microbiome Institute, University College Cork, Cork, Ireland.,School of Microbiology, University College Cork, Cork, Ireland
| | | | - Carthage Moran
- APC Microbiome Institute, University College Cork, Cork, Ireland.,School of Medicine, University College Cork, Cork, Ireland
| | - Donal Sheehan
- APC Microbiome Institute, University College Cork, Cork, Ireland.,School of Medicine, University College Cork, Cork, Ireland
| | - James A Nolan
- APC Microbiome Institute, University College Cork, Cork, Ireland.,School of Microbiology, University College Cork, Cork, Ireland
| | - Colin Hill
- APC Microbiome Institute, University College Cork, Cork, Ireland.,School of Microbiology, University College Cork, Cork, Ireland
| | - Cormac G M Gahan
- APC Microbiome Institute, University College Cork, Cork, Ireland.,School of Microbiology, University College Cork, Cork, Ireland.,School of Pharmacy, University College Cork, Cork, Ireland
| | - Susan A Joyce
- APC Microbiome Institute, University College Cork, Cork, Ireland.,School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
| | - Fergus Shanahan
- APC Microbiome Institute, University College Cork, Cork, Ireland.,School of Medicine, University College Cork, Cork, Ireland
| | - Marcus J Claesson
- APC Microbiome Institute, University College Cork, Cork, Ireland.,School of Microbiology, University College Cork, Cork, Ireland
| |
Collapse
|
98
|
van der Beek CM, Dejong CHC, Troost FJ, Masclee AAM, Lenaerts K. Role of short-chain fatty acids in colonic inflammation, carcinogenesis, and mucosal protection and healing. Nutr Rev 2017; 75:286-305. [PMID: 28402523 DOI: 10.1093/nutrit/nuw067] [Citation(s) in RCA: 224] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Short-chain fatty acids (SCFAs), mainly acetate, propionate, and butyrate, produced by microbial fermentation of undigested food substances are believed to play a beneficial role in human gut health. Short-chain fatty acids influence colonic health through various mechanisms. In vitro and ex vivo studies show that SCFAs have anti-inflammatory and anticarcinogenic effects, play an important role in maintaining metabolic homeostasis in colonocytes, and protect colonocytes from external harm. Animal studies have found substantial positive effects of SCFAs or dietary fiber on colonic disease, but convincing evidence in humans is lacking. Most human intervention trials have been conducted in the context of inflammatory bowel disease. Only a limited number of those trials are of high quality, showing little or no favorable effect of SCFA treatment over placebo. Opportunities for future research include exploring the use of combination therapies with anti-inflammatory drugs, prebiotics, or probiotics; the use of prodrugs in the setting of carcinogenesis; or the direct application of SCFAs to improve mucosal healing after colonic surgery.
Collapse
Affiliation(s)
- Christina M van der Beek
- C.M. van der Beek, C.H.C. Dejong, F.J. Troost, A.A.M. Masclee, and K. Lenaerts are with Top Institute Food and Nutrition, Wageningen, the Netherlands. C.M. van der Beek, C.H.C. Dejong, and K. Lenaerts are with the Department of Surgery, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, the Netherlands. C.H.C. Dejong is with the School for Oncology and Developmental Biology (GROW), Maastricht University Medical Center+, Maastricht, the Netherlands. F.J. Troost and A.A.M. Masclee are with the Department of Internal Medicine, Division of Gastroenterology-Hepatology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - Cornelis H C Dejong
- C.M. van der Beek, C.H.C. Dejong, F.J. Troost, A.A.M. Masclee, and K. Lenaerts are with Top Institute Food and Nutrition, Wageningen, the Netherlands. C.M. van der Beek, C.H.C. Dejong, and K. Lenaerts are with the Department of Surgery, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, the Netherlands. C.H.C. Dejong is with the School for Oncology and Developmental Biology (GROW), Maastricht University Medical Center+, Maastricht, the Netherlands. F.J. Troost and A.A.M. Masclee are with the Department of Internal Medicine, Division of Gastroenterology-Hepatology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - Freddy J Troost
- C.M. van der Beek, C.H.C. Dejong, F.J. Troost, A.A.M. Masclee, and K. Lenaerts are with Top Institute Food and Nutrition, Wageningen, the Netherlands. C.M. van der Beek, C.H.C. Dejong, and K. Lenaerts are with the Department of Surgery, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, the Netherlands. C.H.C. Dejong is with the School for Oncology and Developmental Biology (GROW), Maastricht University Medical Center+, Maastricht, the Netherlands. F.J. Troost and A.A.M. Masclee are with the Department of Internal Medicine, Division of Gastroenterology-Hepatology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - Ad A M Masclee
- C.M. van der Beek, C.H.C. Dejong, F.J. Troost, A.A.M. Masclee, and K. Lenaerts are with Top Institute Food and Nutrition, Wageningen, the Netherlands. C.M. van der Beek, C.H.C. Dejong, and K. Lenaerts are with the Department of Surgery, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, the Netherlands. C.H.C. Dejong is with the School for Oncology and Developmental Biology (GROW), Maastricht University Medical Center+, Maastricht, the Netherlands. F.J. Troost and A.A.M. Masclee are with the Department of Internal Medicine, Division of Gastroenterology-Hepatology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - Kaatje Lenaerts
- C.M. van der Beek, C.H.C. Dejong, F.J. Troost, A.A.M. Masclee, and K. Lenaerts are with Top Institute Food and Nutrition, Wageningen, the Netherlands. C.M. van der Beek, C.H.C. Dejong, and K. Lenaerts are with the Department of Surgery, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, the Netherlands. C.H.C. Dejong is with the School for Oncology and Developmental Biology (GROW), Maastricht University Medical Center+, Maastricht, the Netherlands. F.J. Troost and A.A.M. Masclee are with the Department of Internal Medicine, Division of Gastroenterology-Hepatology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, the Netherlands
| |
Collapse
|
99
|
de Bruyn M, Vermeire S. NOD2 and bacterial recognition as therapeutic targets for Crohn’s disease. Expert Opin Ther Targets 2017; 21:1123-1139. [DOI: 10.1080/14728222.2017.1397627] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Magali de Bruyn
- Translational Research in GastroIntestinal Disorders, Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven, Leuven, Belgium
| | - Séverine Vermeire
- Translational Research in GastroIntestinal Disorders, Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven, Leuven, Belgium
- University Hospitals Leuven, Department of Gastroenterology and Hepatology, Leuven, Belgium
| |
Collapse
|
100
|
Monk JM, Lepp D, Wu W, Pauls KP, Robinson LE, Power KA. Navy and black bean supplementation primes the colonic mucosal microenvironment to improve gut health. J Nutr Biochem 2017; 49:89-100. [PMID: 28915390 DOI: 10.1016/j.jnutbio.2017.08.002] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 07/07/2017] [Accepted: 08/07/2017] [Indexed: 02/06/2023]
Abstract
Common beans (Phaseolus vulgaris L.) are enriched in non-digestible fermentable carbohydrates and phenolic compounds that can modulate the colonic microenvironment (microbiota and host epithelial barrier) to improve gut health. In a comprehensive assessment of the impact of two commonly consumed bean varieties (differing in levels and types of phenolic compounds) within the colonic microenvironment, C57Bl/6 mice were fed diets supplemented with 20% cooked navy bean (NB) or black bean (BB) flours or an isocaloric basal diet control (BD) for 3 weeks. NB and BB similarly altered the fecal microbiota community structure (16S rRNA sequencing) notably by increasing the abundance of carbohydrate fermenting bacteria such as Prevotella, S24-7 and Ruminococcus flavefaciens, which coincided with enhanced short chain fatty acid (SCFA) production (microbial-derived carbohydrate fermentation products) and colonic expression of the SCFA receptors GPR-41/-43/-109a. Both NB and BB enhanced multiple aspects of mucus and epithelial barrier integrity vs. BD including: (i) goblet cell number, crypt mucus content and mucin mRNA expression, (ii) anti-microbial defenses (Reg3γ), (iii) crypt length and epithelial cell proliferation, (iv) apical junctional complex components (occludin, JAM-A, ZO-1 and E-cadherin) mRNA expression and (v) reduced serum endotoxin concentrations. Interestingly, biomarkers of colon barrier integrity (crypt height, mucus content, cell proliferation and goblet cell number) were enhanced in BB vs. NB-fed mice, suggesting added benefits attributable to unique BB components (e.g., phenolics). Overall, NB and BB improved baseline colonic microenvironment function by altering the microbial community structure and activity and promoting colon barrier integrity and function; effects which may prove beneficial in attenuating gut-associated diseases.
Collapse
Affiliation(s)
- Jennifer M Monk
- Guelph Research and Development Centre, Agriculture and Agri-Food Canada, Guelph, ON, Canada, N1G 5C9; Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON, Canada, N1G 2W1
| | - Dion Lepp
- Guelph Research and Development Centre, Agriculture and Agri-Food Canada, Guelph, ON, Canada, N1G 5C9
| | - Wenqing Wu
- Guelph Research and Development Centre, Agriculture and Agri-Food Canada, Guelph, ON, Canada, N1G 5C9
| | - K Peter Pauls
- Department of Plant Agriculture, University of Guelph, Guelph, ON, Canada, N1G 2W1
| | - Lindsay E Robinson
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON, Canada, N1G 2W1
| | - Krista A Power
- Guelph Research and Development Centre, Agriculture and Agri-Food Canada, Guelph, ON, Canada, N1G 5C9; Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON, Canada, N1G 2W1.
| |
Collapse
|