101
|
Sampaio-Maia B, Simões-Silva L, Pestana M, Araujo R, Soares-Silva IJ. The Role of the Gut Microbiome on Chronic Kidney Disease. ADVANCES IN APPLIED MICROBIOLOGY 2016; 96:65-94. [PMID: 27565581 DOI: 10.1016/bs.aambs.2016.06.002] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Chronic kidney disease (CKD) is estimated to affect nearly 500 million people worldwide and cardiovascular (CV) disease is a major cause of death in this population. However, therapeutic interventions targeting traditional CV risks are not effective at lowering the incidence of CV events or at delaying the progression of the disease in CKD patients. In recent years, disturbances of normal gut microbiome were recognized in the pathogenesis of diverse chronic diseases. Gut dysbiosis is being unraveled in CKD and pointed as a nontraditional risk factor for CV risk and CKD progression. The most often reported changes in gut microbiome in CKD are related to the lower levels of Bifidobacteriaceae and Lactobacillaceae and to higher levels of Enterobacteriaceae. Although metagenomics brought us an amplified vision on the microbial world that inhabits the human host, it still lacks the sensitivity to characterize the microbiome up to species level, not revealing alterations that occur within specific genus. Here, we review the current state-of-the-art concerning gut dysbiosis in CKD and its role in pathophysiological mechanisms in CKD, particularly in relation with CV risk. Also, the strategies towards prevention and treatment of gut dysbiosis in CKD progression will be discussed.
Collapse
Affiliation(s)
| | | | - M Pestana
- University of Porto, Porto, Portugal; São João Hospital Center, Porto, EPE, Portugal
| | - R Araujo
- University of Porto, Porto, Portugal; Flinders University, Adelaide, SA, Australia
| | | |
Collapse
|
102
|
Association between the gut microbiota and diet: Fetal life, early childhood, and further life. Nutrition 2015; 32:620-7. [PMID: 26946974 DOI: 10.1016/j.nut.2015.12.037] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2015] [Revised: 11/22/2015] [Accepted: 12/25/2015] [Indexed: 12/22/2022]
Abstract
Gut microbiota establishment and further microbiota shifts are very important for maintaining host health throughout life. There are some factors, including genetics, the mother's health and diet, delivery mode, breast or formula feeding, that may influence the gut microbiota. By the end of approximately the first 3 y of life, the gut microbiota becomes an adult-like stable system. Once established, 60 to 70% of the microbiota composition remains stable throughout life, but 30 to 40% can be altered by changes in the diet and other factors such as physical activity, lifestyle, bacterial infections, and antibiotic or surgical treatment. Diet-related factors that influence the gut microbiota in people of all ages are of great interest. Nutrition may have therapeutic success in gut microbiota correction. This review describes current evidence concerning the links between gut microbiota composition and dietary patterns throughout life.
Collapse
|
103
|
Schwarz EI, Martinez-Lozano Sinues P, Bregy L, Gaisl T, Garcia Gomez D, Gaugg MT, Suter Y, Stebler N, Nussbaumer-Ochsner Y, Bloch KE, Stradling JR, Zenobi R, Kohler M. Effects of CPAP therapy withdrawal on exhaled breath pattern in obstructive sleep apnoea. Thorax 2015; 71:110-7. [DOI: 10.1136/thoraxjnl-2015-207597] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2015] [Accepted: 11/16/2015] [Indexed: 11/04/2022]
|
104
|
Dallas DC, Murray NM, Gan J. Proteolytic Systems in Milk: Perspectives on the Evolutionary Function within the Mammary Gland and the Infant. J Mammary Gland Biol Neoplasia 2015; 20:133-47. [PMID: 26179272 PMCID: PMC4637187 DOI: 10.1007/s10911-015-9334-3] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 07/02/2015] [Indexed: 12/11/2022] Open
Abstract
Milk contains elements of numerous proteolytic systems (zymogens, active proteases, protease inhibitors and protease activators) produced in part from blood, in part by mammary epithelial cells and in part by immune cell secretion. Researchers have examined milk proteases for decades, as they can cause major defects in milk quality and cheese production. Most previous research has examined these proteases with the aim to eliminate or control their actions. However, our recent peptidomics research demonstrates that these milk proteases produce specific peptides in healthy milk and continue to function within the infant's gastrointestinal tract. These findings suggest that milk proteases have an evolutionary function in aiding the infant's digestion or releasing functional peptides. In other words, the mother provides the infant with not only dietary proteins but also the means to digest them. However, proteolysis in the milk is controlled by a balance of protease inhibitors and protease activators so that only a small portion of milk proteins are digested within the mammary gland. This regulation presents a question: If proteolysis is beneficial to the infant, what benefits are gained by preventing complete proteolysis through the presence of protease inhibitors? In addition to summarizing what is known about milk proteolytic systems, we explore possible evolutionary explanations for this proteolytic balance.
Collapse
Affiliation(s)
- David C Dallas
- Department of Food Science and Technology, University of California, Davis, One Shields Avenue, Davis, CA, 95616, USA.
- Foods for Health Institute, University of California, Davis, One Shields Avenue, Davis, CA, 95616, USA.
| | - Niamh M Murray
- Institute of Food and Health, School of Agriculture and Food Science, University College Dublin, Belfield, Dublin, Ireland
| | - Junai Gan
- Department of Food Science and Technology, University of California, Davis, One Shields Avenue, Davis, CA, 95616, USA
- Foods for Health Institute, University of California, Davis, One Shields Avenue, Davis, CA, 95616, USA
| |
Collapse
|
105
|
Ferrario C, Duranti S, Milani C, Mancabelli L, Lugli GA, Turroni F, Mangifesta M, Viappiani A, Ossiprandi MC, van Sinderen D, Ventura M. Exploring Amino Acid Auxotrophy in Bifidobacterium bifidum PRL2010. Front Microbiol 2015; 6:1331. [PMID: 26635786 PMCID: PMC4656816 DOI: 10.3389/fmicb.2015.01331] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 11/12/2015] [Indexed: 01/01/2023] Open
Abstract
The acquisition and assimilation strategies followed by members of the infant gut microbiota to retrieve nitrogen from the gut lumen are still largely unknown. In particular, no information on these metabolic processes is available regarding bifidobacteria, which are among the first microbial colonizers of the human intestine. Here, evaluation of amino acid auxotrophy and prototrophy of Bifidobacterium bifidum, with particular emphasis on B. bifidum strain PRL2010 (LMG S-28692), revealed a putative auxotrophy for cysteine. In addition, we hypothesized that cysteine plays a role in the oxidative stress response in B. bifidum. The use of glutathione as an alternative reduced sulfur compound did not alleviate cysteine auxotrophy of this strain, though it was shown to stimulate expression of the genes involved in cysteine biosynthesis, reminiscent of oxidative stress response. When PRL2010 was grown on a medium containing complex substrates, such as whey proteins or casein hydrolysate, we noticed a distinct growth-promoting effect of these compounds. Transcriptional analysis involving B. bifidum PRL2010 cultivated on whey proteins or casein hydrolysate revealed that the biosynthetic pathways for cysteine and methionine are modulated by the presence of casein hydrolysate. Such findings support the notion that certain complex substrates may act as potential prebiotics for bifidobacteria in their ecological niche.
Collapse
Affiliation(s)
- Chiara Ferrario
- Laboratory of Probiogenomics, Department of Life Sciences, University of Parma Parma, Italy
| | - Sabrina Duranti
- Laboratory of Probiogenomics, Department of Life Sciences, University of Parma Parma, Italy
| | - Christian Milani
- Laboratory of Probiogenomics, Department of Life Sciences, University of Parma Parma, Italy
| | - Leonardo Mancabelli
- Laboratory of Probiogenomics, Department of Life Sciences, University of Parma Parma, Italy
| | - Gabriele A Lugli
- Laboratory of Probiogenomics, Department of Life Sciences, University of Parma Parma, Italy
| | - Francesca Turroni
- Laboratory of Probiogenomics, Department of Life Sciences, University of Parma Parma, Italy
| | | | | | - Maria C Ossiprandi
- Department of Medical-Veterinary Science, University of Parma Parma, Italy
| | - Douwe van Sinderen
- APC Microbiome Institute and School of Microbiology, University College Cork, National University of Ireland Cork, Ireland
| | - Marco Ventura
- Laboratory of Probiogenomics, Department of Life Sciences, University of Parma Parma, Italy
| |
Collapse
|
106
|
Vanhauwaert E, Matthys C, Verdonck L, De Preter V. Low-residue and low-fiber diets in gastrointestinal disease management. Adv Nutr 2015; 6:820-7. [PMID: 26567203 PMCID: PMC4642427 DOI: 10.3945/an.115.009688] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Recently, low-residue diets were removed from the American Academy of Nutrition and Dietetics' Nutrition Care Manual due to the lack of a scientifically accepted quantitative definition and the unavailability of a method to estimate the amount of food residue produced. This narrative review focuses on defining the similarities and/or discrepancies between low-residue and low-fiber diets and on the diagnostic and therapeutic values of these diets in gastrointestinal disease management. Diagnostically, a low-fiber/low-residue diet is used in bowel preparation. A bowel preparation is a cleansing of the intestines of fecal matter and secretions conducted before a diagnostic procedure. Therapeutically, a low-fiber/low-residue diet is part of the treatment of acute relapses in different bowel diseases. The available evidence on low-residue and low-fiber diets is summarized. The main findings showed that within human disease research, the terms "low residue" and "low fiber" are used interchangeably, and information related to the quantity of residue in the diet usually refers to the amount of fiber. Low-fiber/low-residue diets are further explored in both diagnostic and therapeutic situations. On the basis of this literature review, the authors suggest redefining a low-residue diet as a low-fiber diet and to quantitatively define a low-fiber diet as a diet with a maximum of 10 g fiber/d. A low-fiber diet instead of a low-residue diet is recommended as a diagnostic value or as specific therapy for gastrointestinal conditions.
Collapse
Affiliation(s)
- Erika Vanhauwaert
- Knowledge and Innovation Centre Food and Nutrition, University College Leuven-Limburg, Leuven, Belgium; Department of Clinical and Experimental Medicine and
| | - Christophe Matthys
- Department of Clinical and Experimental Medicine and,Clinical Nutrition Unit, Department of Endocrinology, University Hospitals Leuven, Leuven, Belgium
| | - Lies Verdonck
- Knowledge and Innovation Centre Food and Nutrition, University College Leuven-Limburg, Leuven, Belgium
| | - Vicky De Preter
- Knowledge and Innovation Centre Food and Nutrition, University College Leuven-Limburg, Leuven, Belgium;,Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium; and
| |
Collapse
|
107
|
Usami M, Miyoshi M, Yamashita H. Gut microbiota and host metabolism in liver cirrhosis. World J Gastroenterol 2015; 21:11597-11608. [PMID: 26556989 PMCID: PMC4631963 DOI: 10.3748/wjg.v21.i41.11597] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 08/06/2015] [Accepted: 09/30/2015] [Indexed: 02/06/2023] Open
Abstract
The gut microbiota has the capacity to produce a diverse range of compounds that play a major role in regulating the activity of distal organs and the liver is strategically positioned downstream of the gut. Gut microbiota linked compounds such as short chain fatty acids, bile acids, choline metabolites, indole derivatives, vitamins, polyamines, lipids, neurotransmitters and neuroactive compounds, and hypothalamic-pituitary-adrenal axis hormones have many biological functions. This review focuses on the gut microbiota and host metabolism in liver cirrhosis. Dysbiosis in liver cirrhosis causes serious complications, such as bacteremia and hepatic encephalopathy, accompanied by small intestinal bacterial overgrowth and increased intestinal permeability. Gut dysbiosis in cirrhosis and intervention with probiotics and synbiotics in a clinical setting is reviewed and evaluated. Recent studies have revealed the relationship between gut microbiota and host metabolism in chronic metabolic liver disease, especially, non-alcoholic fatty liver disease, alcoholic liver disease, and with the gut microbiota metabolic interactions in dysbiosis related metabolic diseases such as diabetes and obesity. Recently, our understanding of the relationship between the gut and liver and how this regulates systemic metabolic changes in liver cirrhosis has increased. The serum lipid levels of phospholipids, free fatty acids, polyunsaturated fatty acids, especially, eicosapentaenoic acid, arachidonic acid, and docosahexaenoic acid have significant correlations with specific fecal flora in liver cirrhosis. Many clinical and experimental reports support the relationship between fatty acid metabolism and gut-microbiota. Various blood metabolome such as cytokines, amino acids, and vitamins are correlated with gut microbiota in probiotics-treated liver cirrhosis patients. The future evaluation of the gut-microbiota-liver metabolic network and the intervention of these relationships using probiotics, synbiotics, and prebiotics, with sufficient nutrition could aid the development of treatments and prevention for liver cirrhosis patients.
Collapse
|
108
|
Ringel-Kulka T, Choi CH, Temas D, Kim A, Maier DM, Scott K, Galanko JA, Ringel Y. Altered Colonic Bacterial Fermentation as a Potential Pathophysiological Factor in Irritable Bowel Syndrome. Am J Gastroenterol 2015; 110:1339-46. [PMID: 26303129 PMCID: PMC4983766 DOI: 10.1038/ajg.2015.220] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 06/20/2015] [Indexed: 12/11/2022]
Abstract
OBJECTIVES Dysbiosis leading to abnormal intestinal fermentation has been suggested as a possible etiological mechanism in irritable bowel syndrome (IBS). We aimed to investigate the location and magnitude of altered intestinal bacterial fermentation in IBS and its clinical subtypes. METHODS IBS patients who satisfied the Rome III criteria (114) and 33 healthy controls (HC) were investigated. Intestinal fermentation was assessed using two surrogate measures: intestinal intraluminal pH and fecal short-chain fatty acids (SCFAs). Intraluminal pH and intestinal transit times were measured in the small and large bowel using a wireless motility capsule (SmartPill) in 47 IBS and 10 HC. Fecal SCFAs including acetate, propionate, butyrate, and lactate were analyzed by capillary gas chromatography in all enrolled subjects. Correlations between intestinal pH, fecal SCFAs, intestinal transit time, and IBS symptom scores were analyzed. RESULTS Colonic intraluminal pH levels were significantly lower in IBS patients compared with HC (total colonic pH, 6.8 for IBS vs. 7.3 for HC, P=0.042). There were no differences in total and segmental pH levels in the small bowel between IBS patients and HC (6.8 vs. 6.8, P=not significant). The intraluminal colonic pH differences were consistent in all IBS subtypes. Total SCFA level was significantly lower in C-IBS patients than in D-IBS and M-IBS patients and HC. The total SCFA level in all IBS subjects was similar with that of HC. Colonic pH levels correlated positively with colon transit time (CTT) and IBS symptoms severity. Total fecal SCFAs levels correlated negatively with CTT and positively with stool frequency. CONCLUSIONS Colonic intraluminal pH is decreased, suggesting higher colonic fermentation, in IBS patients compared with HC. Fecal SCFAs are not a sensitive marker to estimate intraluminal bacterial fermentation.
Collapse
Affiliation(s)
- Tamar Ringel-Kulka
- Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Chang Hwan Choi
- Division of Gastroenterology and Hepatology, Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Internal Medicine, Chung-Ang University College of Medicine, Seoul, Republic of Korea
| | - Daniel Temas
- Division of Gastroenterology and Hepatology, Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Ari Kim
- Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
- Department of Obstetrics and Gynecology, Institute of Wonkwang Medical Science, Wonkwang University College of Medicine, Iksan, Republic of Korea
| | - Daniele M Maier
- Division of Gastroenterology and Hepatology, Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Karen Scott
- Rowett Institute of Nutrition and Health, University of Aberdeen, Aberdeen, United Kingdom
| | - Joseph A Galanko
- Division of Gastroenterology and Hepatology, Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Yehuda Ringel
- Division of Gastroenterology and Hepatology, Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
109
|
Affiliation(s)
- Aafke W. F. Janssen
- Nutrition, Metabolism, and Genomics Group, Division of Human Nutrition, Wageningen University, Wageningen, The Netherlands
| | - Sander Kersten
- Nutrition, Metabolism, and Genomics Group, Division of Human Nutrition, Wageningen University, Wageningen, The Netherlands
| |
Collapse
|
110
|
Andriamihaja M, Lan A, Beaumont M, Audebert M, Wong X, Yamada K, Yin Y, Tomé D, Carrasco-Pozo C, Gotteland M, Kong X, Blachier F. The deleterious metabolic and genotoxic effects of the bacterial metabolite p-cresol on colonic epithelial cells. Free Radic Biol Med 2015; 85:219-27. [PMID: 25881551 DOI: 10.1016/j.freeradbiomed.2015.04.004] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 03/20/2015] [Accepted: 04/06/2015] [Indexed: 11/21/2022]
Abstract
p-Cresol that is produced by the intestinal microbiota from the amino acid tyrosine is found at millimolar concentrations in the human feces. The effects of this metabolite on colonic epithelial cells were tested in this study. Using the human colonic epithelial HT-29 Glc(-/+) cell line, we found that 0.8mM p-cresol inhibits cell proliferation, an effect concomitant with an accumulation of the cells in the S phase and with a slight increase of cell detachment without necrotic effect. At this concentration, p-cresol inhibited oxygen consumption in HT-29 Glc(-/+) cells. In rat normal colonocytes, p-cresol also inhibited respiration. Pretreatment of HT-29 Glc(-/+) cells with 0.8mM p-cresol for 1 day resulted in an increase of the state 3 oxygen consumption and of the cell maximal respiratory capacity with concomitant increased anion superoxide production. At higher concentrations (1.6 and 3.2mM), p-cresol showed similar effects but additionally increased after 1 day the proton leak through the inner mitochondrial membrane, decreasing the mitochondrial bioenergetic activity. At these concentrations, p-cresol was found to be genotoxic toward HT-29 Glc(-/+) and also LS-174T intestinal cells. Lastly, a decreased ATP intracellular content was observed after 3 days treatment. p-Cresol at 0.8mM concentration inhibits colonocyte respiration and proliferation. In response, cells can mobilize their "respiratory reserve." At higher concentrations, p-cresol pretreatment uncouples cell respiration and ATP synthesis, increases DNA damage, and finally decreases the ATP cell content. Thus, we have identified p-cresol as a metabolic troublemaker and as a genotoxic agent toward colonocytes.
Collapse
Affiliation(s)
- Mireille Andriamihaja
- UMR 914 INRA/AgroParisTech, Nutrition Physiology and Ingestive Behavior, Paris, France
| | - Annaïg Lan
- UMR 914 INRA/AgroParisTech, Nutrition Physiology and Ingestive Behavior, Paris, France
| | - Martin Beaumont
- UMR 914 INRA/AgroParisTech, Nutrition Physiology and Ingestive Behavior, Paris, France
| | - Marc Audebert
- INRA, UMR1331, Toxalim, Research Centre in Food Toxicology, F-31027 Toulouse, France
| | - Ximena Wong
- Department of Nutrition, Faculty of Medicine University of Chile, Santiago, Chile
| | - Kana Yamada
- UMR 914 INRA/AgroParisTech, Nutrition Physiology and Ingestive Behavior, Paris, France
| | - Yulong Yin
- Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| | - Daniel Tomé
- UMR 914 INRA/AgroParisTech, Nutrition Physiology and Ingestive Behavior, Paris, France
| | | | - Martin Gotteland
- Department of Nutrition, Faculty of Medicine University of Chile, Santiago, Chile; Institute of Nutrition and Food Technology (INTA), University of Chile, Santiago, Chile
| | - Xiangfeng Kong
- Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| | - François Blachier
- UMR 914 INRA/AgroParisTech, Nutrition Physiology and Ingestive Behavior, Paris, France.
| |
Collapse
|
111
|
Identification of metabolic signatures linked to anti-inflammatory effects of Faecalibacterium prausnitzii. mBio 2015; 6:mBio.00300-15. [PMID: 25900655 PMCID: PMC4453580 DOI: 10.1128/mbio.00300-15] [Citation(s) in RCA: 185] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
UNLABELLED Faecalibacterium prausnitzii is an anti-inflammatory commensal bacterium identified on the basis of human clinical data. The mechanisms underlying its beneficial effects are still unknown. Gnotobiotic mice harboring F. prausnitzii (A2-165) and Escherichia coli (K-12 JM105) were subjected to 2,4,6-trinitrobenzenesulfonic acid (TNBS)-induced acute colitis. The inflammatory colitis scores and a gas chromatography-time of flight (GC/TOF) mass spectrometry-based metabolomic profile were monitored in blood, ileum, cecum, colon, and feces in gnotobiotic mice. The potential anti-inflammatory metabolites were tested in vitro. We obtained stable E. coli and F. prausnitzii-diassociated mice in which E. coli primed the gastrointestinal tract (GIT), allowing a durable and stable establishment of F. prausnitzii. The disease activity index, histological scores, myeloperoxidase (MPO) activity, and serum cytokine levels were significantly lower in the presence of F. prausnitzii after TNBS challenge. The protective effect of F. prausnitzii against colitis was correlated to its implantation level and was linked to overrepresented metabolites along the GIT and in serum. Among 983 metabolites in GIT samples and serum, 279 were assigned to known chemical reactions. Some of them, belonging to the ammonia (α-ketoglutarate), osmoprotective (raffinose), and phenolic (including anti-inflammatory shikimic and salicylic acids) pathways, were associated with a protective effect of F. prausnitzii, and the functional link was established in vitro for salicylic acid. We show for the first time that F. prausnitzii is a highly active commensal bacterium involved in reduction of colitis through in vivo modulation of metabolites along the GIT and in the peripheral blood. IMPORTANCE Inflammatory bowel diseases (IBD) are characterized by low proportions of F. prausnitzii in the gut microbiome. This commensal bacterium exhibits anti-inflammatory effects through still unknown mechanisms. Stable monoassociated rodents are actually not a reproducible model to decipher F. prausnitzii protective effects. We propose a new gnotobiotic rodent model providing mechanistic clues. In this model, F. prausnitzii exhibits protective effects against an acute colitis and a protective metabolic profile is linked to its presence along the digestive tract. We identified a molecule, salicylic acid, directly involved in the protective effect of F. prausnitzii. Targeting its metabolic pathways could be an attractive therapeutic strategy in IBD.
Collapse
|
112
|
De Preter V, Machiels K, Joossens M, Arijs I, Matthys C, Vermeire S, Rutgeerts P, Verbeke K. Faecal metabolite profiling identifies medium-chain fatty acids as discriminating compounds in IBD. Gut 2015; 64:447-58. [PMID: 24811995 DOI: 10.1136/gutjnl-2013-306423] [Citation(s) in RCA: 163] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Bacteria play a role in the onset and perpetuation of intestinal inflammation in IBD. Compositional alterations may also change the metabolic capacities of the gut bacteria. OBJECTIVE To examine the metabolic activity of the microbiota of patients with Crohn's disease (CD), UC or pouchitis compared with healthy controls (HC) and determine whether eventual differences might be related to the pathogenesis of the disease. METHODS Faecal samples were obtained from 40 HC, 83 patients with CD, 68 with UC and 13 with pouchitis. Disease activity was assessed in CD using the Harvey-Bradshaw Index, in UC using the UC Disease Activity Index and in pouchitis using the Pouchitis Disease Activity Index. Metabolite profiles were analysed using gas chromatography-mass spectrometry. RESULTS The number of metabolites identified in HC (54) was significantly higher than in patients with CD (44, p<0.001), UC (47, p=0.042) and pouchitis (43, p=0.036). Multivariate discriminant analysis predicted HC, CD, UC and pouchitis group membership with high sensitivity and specificity. The levels of medium-chain fatty acids (MCFAs: pentanoate, hexanoate, heptanoate, octanoate and nonanoate), and of some protein fermentation metabolites, were significantly decreased in patients with CD, UC and pouchitis. Hexanoate levels were inversely correlated to disease activity in CD (correlation coefficient=-0.157, p=0.046), whereas a significant positive correlation was found between styrene levels and disease activity in UC (correlation coefficient=0.338, p=0.001). CONCLUSIONS Faecal metabolic profiling in patients with IBD relative to healthy controls identified MCFAs as important metabolic biomarkers of disease-related changes. TRIAL REGISTRATION NO NCT 01666717.
Collapse
Affiliation(s)
- Vicky De Preter
- Translational Research Center for Gastrointestinal Disorders (TARGID) and Leuven Food Science and Nutrition Research Centre (LFoRCe), University Hospital Gasthuisberg, KU Leuven, Leuven, Belgium
| | - Kathleen Machiels
- Translational Research Center for Gastrointestinal Disorders (TARGID) and Leuven Food Science and Nutrition Research Centre (LFoRCe), University Hospital Gasthuisberg, KU Leuven, Leuven, Belgium
| | - Marie Joossens
- Translational Research Center for Gastrointestinal Disorders (TARGID) and Leuven Food Science and Nutrition Research Centre (LFoRCe), University Hospital Gasthuisberg, KU Leuven, Leuven, Belgium Department of Structural Biology, Research group of Bioinformatics and (Eco-)Systems Biology, VIB, Brussels, Belgium Microbiology Unit (MICR), Department of Applied Biological Sciences (DBIT), Vrije Universiteit Brussel, Brussels, Belgium
| | - Ingrid Arijs
- Translational Research Center for Gastrointestinal Disorders (TARGID) and Leuven Food Science and Nutrition Research Centre (LFoRCe), University Hospital Gasthuisberg, KU Leuven, Leuven, Belgium
| | - Christophe Matthys
- Department of Clinical and Experimental Endocrinology, Subdivision Clinical Nutrition, University Hospital Gasthuisberg, KULeuven, Leuven, Belgium
| | - Severine Vermeire
- Translational Research Center for Gastrointestinal Disorders (TARGID) and Leuven Food Science and Nutrition Research Centre (LFoRCe), University Hospital Gasthuisberg, KU Leuven, Leuven, Belgium
| | - Paul Rutgeerts
- Translational Research Center for Gastrointestinal Disorders (TARGID) and Leuven Food Science and Nutrition Research Centre (LFoRCe), University Hospital Gasthuisberg, KU Leuven, Leuven, Belgium
| | - Kristin Verbeke
- Translational Research Center for Gastrointestinal Disorders (TARGID) and Leuven Food Science and Nutrition Research Centre (LFoRCe), University Hospital Gasthuisberg, KU Leuven, Leuven, Belgium
| |
Collapse
|
113
|
Down-regulation of monocarboxylate transporter 1 (MCT1) gene expression in the colon of piglets is linked to bacterial protein fermentation and pro-inflammatory cytokine-mediated signalling. Br J Nutr 2015; 113:610-7. [PMID: 25656974 DOI: 10.1017/s0007114514004231] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The present study investigated the influence of bacterial metabolites on monocarboxylate transporter 1 (MCT1) expression in pigs using in vivo, ex vivo and in vitro approaches. Piglets (n 24) were fed high-protein (26 %) or low-protein (18 %) diets with or without fermentable carbohydrates. Colonic digesta samples were analysed for a broad range of bacterial metabolites. The expression of MCT1, TNF-α, interferon γ (IFN-γ) and IL-8 was determined in colonic tissue. The expression of MCT1 was lower and of TNF-α and IL-8 was higher with high-protein diets (P< 0·05). MCT1 expression was positively correlated with l-lactate, whereas negatively correlated with NH₃ and putrescine (P< 0·05). The expression of IL-8 and TNF-α was negatively correlated with l-lactate and positively correlated with NH₃ and putrescine, whereas the expression of IFN-γ was positively correlated with histamine and 4-ethylphenol (P< 0·05). Subsequently, porcine colonic tissue and Caco-2 cells were incubated with Na-butyrate, NH₄Cl or TNF-α as selected bacterial metabolites or mediators of inflammation. Colonic MCT1 expression was higher after incubation with Na-butyrate (P< 0·05) and lower after incubation with NH₄Cl or TNF-α (P< 0·05). Incubation of Caco-2 cells with increasing concentrations of these metabolites confirmed the up-regulation of MCT1 expression by Na-butyrate (linear, P< 0·05) and down-regulation by TNF-α and NH₄Cl (linear, P< 0·05). The high-protein diet decreased the expression of MCT1 in the colon of pigs, which appears to be linked to NH₃- and TNF-α-mediated signalling.
Collapse
|
114
|
Andersen V, Vogel U. Interactions between meat intake and genetic variation in relation to colorectal cancer. GENES AND NUTRITION 2014; 10:448. [PMID: 25491747 PMCID: PMC4261072 DOI: 10.1007/s12263-014-0448-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2014] [Accepted: 11/28/2014] [Indexed: 01/03/2023]
Abstract
Meat intake is associated with the risk of colorectal cancer. The objective of this systematic review was to evaluate interactions between meat intake and genetic variation in order to identify biological pathways involved in meat carcinogenesis. We performed a literature search of PubMed and Embase using “interaction”, “meat”, “polymorphisms”, and “colorectal cancer”, and data on meat–gene interactions were extracted. The studies were divided according to whether information on meat intake was collected prospectively or retrospectively. In prospective studies, interactions between meat intake and polymorphisms in PTGS2 (encoding COX-2), ABCB1, IL10, NFKB1, MSH3, XPC (Pint = 0.006, 0.01, 0.04, 0.03, 0.002, 0.01, respectively), but not IL1B, HMOX1, ABCC2, ABCG2, NR1I2 (encoding PXR), NR1H2 (encoding LXR), NAT1, NAT2, MSH6, or MLH1 in relation to CRC were found. Interaction between a polymorphism in XPC and meat was found in one prospective and one case–control study; however, the directions of the risk estimates were opposite. Thus, none of the findings were replicated. The results from this systematic review suggest that genetic variation in the inflammatory response and DNA repair pathway is involved in meat-related colorectal carcinogenesis, whereas no support for the involvement of heme and iron from meat or cooking mutagens was found. Further studies assessing interactions between meat intake and genetic variation in relation to CRC in large well-characterised prospective cohorts with relevant meat exposure are warranted.
Collapse
Affiliation(s)
- Vibeke Andersen
- Organ Center, Hospital of Southern Jutland, Aabenraa, Denmark,
| | | |
Collapse
|
115
|
Bertolotti M, Lonardo A, Mussi C, Baldelli E, Pellegrini E, Ballestri S, Romagnoli D, Loria P. Nonalcoholic fatty liver disease and aging: Epidemiology to management. World J Gastroenterol 2014; 20:14185-14204. [PMID: 25339806 PMCID: PMC4202348 DOI: 10.3748/wjg.v20.i39.14185] [Citation(s) in RCA: 208] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Revised: 02/17/2014] [Accepted: 06/17/2014] [Indexed: 02/06/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is common in the elderly, in whom it carries a more substantial burden of hepatic (nonalcoholic steatohepatitis, cirrhosis and hepatocellular carcinoma) and extra-hepatic manifestations and complications (cardiovascular disease, extrahepatic neoplasms) than in younger age groups. Therefore, proper identification and management of this condition is a major task for clinical geriatricians and geriatric hepatologists. In this paper, the epidemiology and pathophysiology of this condition are reviewed, and a full discussion of the link between NAFLD and the aspects that are peculiar to elderly individuals is provided; these aspects include frailty, multimorbidity, polypharmacy and dementia. The proper treatment strategy will have to consider the peculiarities of geriatric patients, so a multidisciplinary approach is mandatory. Non-pharmacological treatment (diet and physical exercise) has to be tailored individually considering the physical limitations of most elderly people and the need for an adequate caloric supply. Similarly, the choice of drug treatment must carefully balance the benefits and risks in terms of adverse events and pharmacological interactions in the common context of both multiple health conditions and polypharmacy. In conclusion, further epidemiological and pathophysiological insight is warranted. More accurate understanding of the molecular mechanisms of geriatric NAFLD will help in identifying the most appropriate diagnostic and therapeutic approach for individual elderly patients.
Collapse
|
116
|
Intestinal permeability, gut-bacterial dysbiosis, and behavioral markers of alcohol-dependence severity. Proc Natl Acad Sci U S A 2014; 111:E4485-93. [PMID: 25288760 DOI: 10.1073/pnas.1415174111] [Citation(s) in RCA: 622] [Impact Index Per Article: 62.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Alcohol dependence has traditionally been considered a brain disorder. Alteration in the composition of the gut microbiota has recently been shown to be present in psychiatric disorders, which suggests the possibility of gut-to-brain interactions in the development of alcohol dependence. The aim of the present study was to explore whether changes in gut permeability are linked to gut-microbiota composition and activity in alcohol-dependent subjects. We also investigated whether gut dysfunction is associated with the psychological symptoms of alcohol dependence. Finally, we tested the reversibility of the biological and behavioral parameters after a short-term detoxification program. We found that some, but not all, alcohol-dependent subjects developed gut leakiness, which was associated with higher scores of depression, anxiety, and alcohol craving after 3 wk of abstinence, which may be important psychological factors of relapse. Moreover, subjects with increased gut permeability also had altered composition and activity of the gut microbiota. These results suggest the existence of a gut-brain axis in alcohol dependence, which implicates the gut microbiota as an actor in the gut barrier and in behavioral disorders. Thus, the gut microbiota seems to be a previously unidentified target in the management of alcohol dependence.
Collapse
|
117
|
Toivonen RK, Emani R, Munukka E, Rintala A, Laiho A, Pietilä S, Pursiheimo JP, Soidinsalo P, Linhala M, Eerola E, Huovinen P, Hänninen A. Fermentable fibres condition colon microbiota and promote diabetogenesis in NOD mice. Diabetologia 2014; 57:2183-92. [PMID: 25031069 DOI: 10.1007/s00125-014-3325-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Accepted: 06/17/2014] [Indexed: 12/16/2022]
Abstract
AIMS/HYPOTHESIS Gut microbiota (GM) and diet both appear to be important in the pathogenesis of type 1 diabetes. Fermentable fibres (FFs), of which there is an ample supply in natural, diabetes-promoting diets, are used by GM as a source of energy. Our aim was to determine whether FFs modify GM and diabetes incidence in the NOD mouse. METHODS Female NOD mice were weaned to a semisynthetic diet and the effects of FF supplementation on diabetes incidence and insulitis were evaluated. Real-time quantitative PCR was employed to determine the effects imposed to gene transcripts in the colon and lymph nodes. Changes to GM were analysed by next-generation sequencing. RESULTS NOD mice fed semisynthetic diets free from FFs were largely protected from diabetes while semisynthetic diets supplemented with the FFs pectin and xylan (PX) resulted in higher diabetes incidence. Semisynthetic diet free from FFs altered GM composition significantly; addition of PX changed the composition of the GM towards that found in natural-diet-fed mice and increased production of FF-derived short-chain fatty acid metabolites in the colon. The highly diabetogenic natural diet was associated with expression of proinflammatory and stress-related genes in the colon, while the semisynthetic diet free from FFs promoted Il4, Il22, Tgfβ and Foxp3 transcripts in the colon and/or pancreatic lymph node. PX in the same diet counteracted these effects and promoted stress-related IL-18 activation in gut epithelial cells. 16S RNA sequencing revealed each diet to give rise to its particular GM composition, with different Firmicutes to Bacteroidetes ratios, and enrichment of mucin-degrading Ruminococcaceae following diabetes-protective FF-free diet. CONCLUSIONS/INTERPRETATION FFs condition microbiota, affect colon homeostasis and are important components of natural, diabetes-promoting diets in NOD mice.
Collapse
Affiliation(s)
- Raine K Toivonen
- Department of Medical Microbiology and Immunology, University of Turku, Kiinamyllynkatu 13, 20520, Turku, Finland,
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
118
|
Abstract
A large body of evidence suggests that the regulation of energy balance and glucose homeostasis by fermentable carbohydrates induces specific changes in the gut microbiota. Among the mechanisms, our research group and others have demonstrated that the gut microbiota fermentation (i.e., bacterial digestion of specific compounds) of specific prebiotics or other non-digestible carbohydrates is associated with the secretion of enteroendocrine peptides, such as the glucagon-like peptide-1 (GLP-1) and peptide YY (PYY), produced by L-cells. In this review, we highlight past and recent results describing how dietary manipulation of the gut microbiota, using nutrients or specific microbes, can stimulate GLP-1 secretion in rodents and humans. Furthermore, the purpose of this review is to discuss the putative mechanisms by which specific bacterial metabolites, such as short chain fatty acids, trigger GLP-1 secretion through GPR41/43-dependent mechanisms. Moreover, we conclude by discussing the molecular advance showing that the endocannabinoid system or related bioactive lipids modulated by the gut microbiota may contribute to the regulation of glucose, lipid and energy homeostasis.
Collapse
Affiliation(s)
- Amandine Everard
- WELBIO (Walloon Excellence in Life sciences and BIOtechnology), Metabolism and Nutrition research group, Université catholique de Louvain, Louvain Drug Research Institute, Av. E. Mounier, 73, Box B1.73.11, 1200, Brussels, Belgium
| | | |
Collapse
|
119
|
Solga SF. Breath volatile organic compounds for the gut-fatty liver axis: Promise, peril, and path forward. World J Gastroenterol 2014; 20:9017-9025. [PMID: 25083075 PMCID: PMC4112861 DOI: 10.3748/wjg.v20.i27.9017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Revised: 01/15/2014] [Accepted: 04/09/2014] [Indexed: 02/06/2023] Open
Abstract
The worldwide interest in the gut microbiome and its impact on the upstream liver highlight a critical upside to breath research: it can uniquely measure otherwise unmeasurable biology. Bacteria make gases [volatile organic compounds (VOCs)] that are directly relevant to pathophysiology of the fatty liver and associated conditions, including obesity. Measurement of these VOCs and their metabolites in the exhaled breath, therefore, present an opportunity to safely and easily evaluate, on both a personal and a population level, some of our most pressing public health threats. This is an opportunity that must be pursued. To date, however, breath analysis remains a slowly evolving field which only occasionally impacts clinical research or patient care. One major obstacle to progress is that breath analysis is inherently and emphatically mutli-disciplinary: it connects engineering, chemistry, breath mechanics, biology and medicine. Unbalanced or incomplete teams may produce inconsistent and often unsatisfactory results. A second impediment is the lack of a well-known stepwise structure for the development of non-invasive diagnostics. As a result, the breath research landscape is replete with orphaned single-center pilot studies. Often, important hypotheses and key observations have not been pursued to maturation. This paper reviews the rationale and requirements for breath VOC research applied to the gut-fatty liver axis and offers some suggestions for future development.
Collapse
|
120
|
Goedert JJ, Sampson JN, Moore SC, Xiao Q, Xiong X, Hayes RB, Ahn J, Shi J, Sinha R. Fecal metabolomics: assay performance and association with colorectal cancer. Carcinogenesis 2014; 35:2089-96. [PMID: 25037050 DOI: 10.1093/carcin/bgu131] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Metabolomic analysis of feces may provide insights on colorectal cancer (CRC) if assay performance is satisfactory. In lyophilized feces from 48 CRC cases, 102 matched controls, and 48 masked quality control specimens, 1043 small molecules were detected with a commercial platform. Assay reproducibility was good for 527 metabolites [technical intraclass correlation coefficient (ICC) >0.7 in quality control specimens], but reproducibility in 6-month paired specimens was lower for the majority of metabolites (within-subject ICC ≤0.5). In the CRC cases and controls, significant differences (false discovery rate ≤0.10) were found for 41 of 1043 fecal metabolites. Direct cancer association was found with three fecal heme-related molecules [covariate-adjusted 90th versus 10th percentile odds ratio (OR) = 17-345], 18 peptides/amino acids (OR = 3-14), palmitoyl-sphingomyelin (OR = 14), mandelate (OR = 3) and p-hydroxy-benzaldehyde (OR = 4). Conversely, cancer association was inverse with acetaminophen metabolites (OR <0.1), tocopherols (OR = 0.3), sitostanol (OR = 0.2), 3-dehydrocarnitine (OR = 0.4), pterin (OR = 0.3), conjugated-linoleate-18-2N7 (OR = 0.2), N-2-furoyl-glycine (OR = 0.3) and p-aminobenzoate (PABA, OR = 0.2). Correlations suggested an independent role for palmitoyl-sphingomyelin and a central role for PABA (which was stable over 6 months, within-subject ICC 0.67) modulated by p-hydroxy-benzaldehyde. Power calculations based on ICCs indicate that only 45% of metabolites with a true relative risk 5.0 would be found in prospectively collected, prediagnostic specimens from 500 cases and 500 controls. Thus, because fecal metabolites vary over time, very large studies will be needed to reliably detect associations of many metabolites that potentially contribute to CRC.
Collapse
Affiliation(s)
- James J Goedert
- Epidemiology and Biostatistics Program, Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9609 Medical Center Drive, Bethesda, MD 20892-9704, USA, Information Management Services, 6110 Executive Boulevard, Rockville, MD 20852, USA and Division of Epidemiology, Department of Population Health, New York University School of Medicine, 650 First Avenue, #518, New York, NY 10016, USA
| | - Joshua N Sampson
- Epidemiology and Biostatistics Program, Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9609 Medical Center Drive, Bethesda, MD 20892-9704, USA, Information Management Services, 6110 Executive Boulevard, Rockville, MD 20852, USA and Division of Epidemiology, Department of Population Health, New York University School of Medicine, 650 First Avenue, #518, New York, NY 10016, USA
| | - Steven C Moore
- Epidemiology and Biostatistics Program, Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9609 Medical Center Drive, Bethesda, MD 20892-9704, USA, Information Management Services, 6110 Executive Boulevard, Rockville, MD 20852, USA and Division of Epidemiology, Department of Population Health, New York University School of Medicine, 650 First Avenue, #518, New York, NY 10016, USA
| | - Qian Xiao
- Epidemiology and Biostatistics Program, Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9609 Medical Center Drive, Bethesda, MD 20892-9704, USA, Information Management Services, 6110 Executive Boulevard, Rockville, MD 20852, USA and Division of Epidemiology, Department of Population Health, New York University School of Medicine, 650 First Avenue, #518, New York, NY 10016, USA
| | - Xiaoqin Xiong
- Information Management Services, 6110 Executive Boulevard, Rockville, MD 20852, USA and
| | - Richard B Hayes
- Division of Epidemiology, Department of Population Health, New York University School of Medicine, 650 First Avenue, #518, New York, NY 10016, USA
| | - Jiyoung Ahn
- Division of Epidemiology, Department of Population Health, New York University School of Medicine, 650 First Avenue, #518, New York, NY 10016, USA
| | - Jianxin Shi
- Epidemiology and Biostatistics Program, Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9609 Medical Center Drive, Bethesda, MD 20892-9704, USA, Information Management Services, 6110 Executive Boulevard, Rockville, MD 20852, USA and Division of Epidemiology, Department of Population Health, New York University School of Medicine, 650 First Avenue, #518, New York, NY 10016, USA
| | - Rashmi Sinha
- Epidemiology and Biostatistics Program, Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9609 Medical Center Drive, Bethesda, MD 20892-9704, USA, Information Management Services, 6110 Executive Boulevard, Rockville, MD 20852, USA and Division of Epidemiology, Department of Population Health, New York University School of Medicine, 650 First Avenue, #518, New York, NY 10016, USA
| |
Collapse
|
121
|
Pieper R, Boudry C, Bindelle J, Vahjen W, Zentek J. Interaction between dietary protein content and the source of carbohydrates along the gastrointestinal tract of weaned piglets. Arch Anim Nutr 2014; 68:263-80. [PMID: 24979393 DOI: 10.1080/1745039x.2014.932962] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Although fermentable carbohydrates (CHO) can reduce metabolites derived from dietary protein fermentation in the intestine of pigs, the interaction between site of fermentation and substrate availability along the gut is still unclear. The current study aimed at determining the impact of two different sources of carbohydrates in diets with low or very high protein content on microbial metabolite profiles along the gastrointestinal tract of piglets. Thirty-six piglets (n = 6 per group) were fed diets high (26%, HP) or low (18%, LP) in dietary protein and with or without two different sources of carbohydrates (12% sugar beet pulp, SBP, or 8% lignocellulose, LNC) in a 2 × 3 factorial design. After 3 weeks, contents from stomach, jejunum, ileum, caecum, proximal and distal colon were taken and analysed for major bacterial metabolites (D-lactate, L-lactate, short chain fatty acids, ammonia, amines, phenols and indols). Results indicate considerable fermentation of CHO and protein already in the stomach. HP diets increased the formation of ammonia, amines, phenolic and indolic compounds throughout the different parts of the intestine with most pronounced effects in the distal colon. Dietary SBP inclusion in LP diets favoured the formation of cadaverine in the proximal parts of the intestine. SBP mainly increased CHO-derived metabolites such as SCFA and lactate and decreased protein-derived metabolites in the large intestine. Based on metabolite profiles, LNC was partly fermented in the distal large intestine and reduced mainly phenols, indols and cadaverine, but not ammonia. Multivariate analysis confirmed more diet-specific metabolite patterns in the stomach, whereas the CHO addition was the main determinant in the caecum and proximal colon. The protein level mainly influenced the metabolite patterns in the distal colon. The results confirm the importance of CHO source to influence the formation of metabolites derived from protein fermentation along the intestinal tract of the pig.
Collapse
Affiliation(s)
- Robert Pieper
- a Department of Veterinary Medicine , Institute of Animal Nutrition, Freie Universität Berlin , Berlin , Germany
| | | | | | | | | |
Collapse
|
122
|
Li L, Somerset S. The clinical significance of the gut microbiota in cystic fibrosis and the potential for dietary therapies. Clin Nutr 2014; 33:571-80. [PMID: 24767984 DOI: 10.1016/j.clnu.2014.04.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Revised: 04/03/2014] [Accepted: 04/04/2014] [Indexed: 02/08/2023]
Abstract
Cystic fibrosis (CF) is characterised by many comorbidities related to aberrant mucosa and chronic inflammation in the respiratory and digestive systems. The intestinal mucosa serves as the primary interface between the gut microbiota and endocrine, neural and immune systems. There is emerging evidence that aberrant intestinal mucosa in CF may associate with an altered gut microbiota. Compared to healthy subjects, the overall bacterial abundance and species richness seems to be reduced in CF, accompanied by a trend in suppression of Firmicutes and Bacteroidetes spp. and an augmentation of potentially pathogenic species. There is also some concordance of gut and respiratory microbiotas in CF infants over time. The clinical significance of these observations awaits investigation. The gut microbiota have some potential in CF management by affecting inflammatory and immune responses, and influencing aberrant mucosa. As an important modifiable factor, diet therapies such as probiotics and prebiotics have shown initial promise in improving CF related conditions associated with chronic inflammation. More studies are needed to confirm this, as well as the efficacy of other dietary strategies such as modulating dietary fat and indigestible carbohydrate. Similarly, dietary modification of gut microbiota to optimise nutritional status in CF may be feasible, although more CF-specific studies are warranted.
Collapse
Affiliation(s)
- Li Li
- School of Public Health, Griffith Health Institute, Griffith University, Brisbane, Queensland, Australia.
| | - Shawn Somerset
- School of Allied Health, Australian Catholic University, PO Box 456, Virginia, Brisbane, Queensland 4014, Australia.
| |
Collapse
|
123
|
Shoaie S, Nielsen J. Elucidating the interactions between the human gut microbiota and its host through metabolic modeling. Front Genet 2014; 5:86. [PMID: 24795748 PMCID: PMC4000997 DOI: 10.3389/fgene.2014.00086] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Accepted: 03/31/2014] [Indexed: 01/03/2023] Open
Abstract
Increased understanding of the interactions between the gut microbiota, diet and environmental effects may allow us to design efficient treatment strategies for addressing global health problems. Existence of symbiotic microorganisms in the human gut provides different functions for the host such as conversion of nutrients, training of the immune system, and resistance to pathogens. The gut microbiome also plays an influential role in maintaining human health, and it is a potential target for prevention and treatment of common disorders including obesity, type 2 diabetes, and atherosclerosis. Due to the extreme complexity of such disorders, it is necessary to develop mathematical models for deciphering the role of its individual elements as well as the entire system and such models may assist in better understanding of the interactions between the bacteria in the human gut and the host by use of genome-scale metabolic models (GEMs). Recently, GEMs have been employed to explore the interactions between predominant bacteria in the gut ecosystems. Additionally, these models enabled analysis of the contribution of each species to the overall metabolism of the microbiota through the integration of omics data. The outcome of these studies can be used for proposing optimal conditions for desired microbiome phenotypes. Here, we review the recent progress and challenges for elucidating the interactions between the human gut microbiota and host through metabolic modeling. We discuss how these models may provide scaffolds for analyzing high-throughput data, developing probiotics and prebiotics, evaluating the effects of probiotics and prebiotics and eventually designing clinical interventions.
Collapse
Affiliation(s)
- Saeed Shoaie
- Department of Chemical and Biological Engineering, Chalmers University of Technology Gothenburg, Sweden
| | - Jens Nielsen
- Department of Chemical and Biological Engineering, Chalmers University of Technology Gothenburg, Sweden
| |
Collapse
|
124
|
Degirolamo C, Rainaldi S, Bovenga F, Murzilli S, Moschetta A. Microbiota Modification with Probiotics Induces Hepatic Bile Acid Synthesis via Downregulation of the Fxr-Fgf15 Axis in Mice. Cell Rep 2014; 7:12-8. [DOI: 10.1016/j.celrep.2014.02.032] [Citation(s) in RCA: 233] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Revised: 01/16/2014] [Accepted: 02/22/2014] [Indexed: 02/06/2023] Open
|
125
|
Walsh CJ, Guinane CM, O'Toole PW, Cotter PD. Beneficial modulation of the gut microbiota. FEBS Lett 2014; 588:4120-30. [PMID: 24681100 DOI: 10.1016/j.febslet.2014.03.035] [Citation(s) in RCA: 173] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 03/14/2014] [Accepted: 03/17/2014] [Indexed: 12/21/2022]
Abstract
The human gut microbiota comprises approximately 100 trillion microbial cells and has a significant effect on many aspects of human physiology including metabolism, nutrient absorption and immune function. Disruption of this population has been implicated in many conditions and diseases, including examples such as obesity, inflammatory bowel disease and colorectal cancer that are highlighted in this review. A logical extension of these observations suggests that the manipulation of the gut microbiota can be employed to prevent or treat these conditions. Thus, here we highlight a variety of options, including the use of changes in diet (including the use of prebiotics), antimicrobial-based intervention, probiotics and faecal microbiota transplantation, and discuss their relative merits with respect to modulating the intestinal community in a beneficial way.
Collapse
Affiliation(s)
- Calum J Walsh
- Teagasc Food Research Centre, Moorepark, Fermoy, County Cork, Ireland; Department of Microbiology, University College Cork, Cork, Ireland
| | | | - Paul W O'Toole
- Department of Microbiology, University College Cork, Cork, Ireland; Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland
| | - Paul D Cotter
- Teagasc Food Research Centre, Moorepark, Fermoy, County Cork, Ireland; Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland.
| |
Collapse
|
126
|
Abstract
The etiology of colon cancer is complex, yet it is undoubtedly impacted by intestinal microbiota. Whether the contribution to colon carcinogenesis is generated through the presence of an overall dysbiosis or by specific pathogens is still a matter for debate. However, it is apparent that interactions between microbiota and the host are mediated by a variety of processes, including signaling cascades, the immune system, host metabolism, and regulation of gene transcription. To fully appreciate the role of microbiota in colon carcinogenesis, it will be necessary to expand efforts to define populations in niche environments, such as colonic crypts, explore cross talk between the host and the microbiota, and more completely define the metabolomic profile of the microbiota. These efforts must be pursued with appreciation that dietary substrates and other environmental modifiers mediate changes in the microbiota, as well as their metabolism and functional characteristics.
Collapse
|
127
|
Gao RY, Zhu QC, Wu W, Qin HL. Compositional differences in fecal microbiota between rats with colorectal cancer and normal rats. Shijie Huaren Xiaohua Zazhi 2014; 22:661-667. [DOI: 10.11569/wcjd.v22.i5.661] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the compositional differences in fecal flora between rats with colorectal cancer and normal rats.
METHODS: A rat model of colorectal cancer was developed by intraperitoneal injection of 1, 2-dimethyl hydrazine (DMH). Fecal samples were collected from rats with colorectal cancer and normal controls, and the microbiota was isolated by PCR-DGGE technique to perform flora similarity analysis (cluster analysis) and polymorphism analysis (richness, uniformity, Shannon-Wiener index, Simpson index) and to compare with the GenBank to identify the genus so as to study the variation.
RESULTS: Compared with normal rats, the abundance of Lachnospiraceae, Ruminococcaceae, Lactobacillus intestinalis, Paraprevotella, Lactobacillus murinus, Lactobacillus, Prevotella, Lactobacillus crispatus and Lachnospiracea incertae sedis was significantly reduced and that of Coprobacillus was significantly increased in rats with colorectal cancer. Although the flora diversity between the two groups showed no statistical difference, there was a significant difference in flora composition.
CONCLUSION: The composition of fecal microflora changes in rats with colorectal cancer compared with normal rats, with the number of beneficial bacteria reduced and that of potential pathogens increased.
Collapse
|
128
|
Nowak A, Śliżewska K. β-Glucuronidase and β-glucosidase activity and human fecal water genotoxicity in the presence of probiotic lactobacilli and the heterocyclic aromatic amine IQ in vitro. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2014; 37:66-73. [PMID: 24309132 DOI: 10.1016/j.etap.2013.10.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Revised: 10/20/2013] [Accepted: 10/24/2013] [Indexed: 06/02/2023]
Abstract
The aim of the study was to assess the genotoxicity of fecal water (FW) and the activity of fecal enzymes (β-glucuronidase and β-glucosidase) after incubation with 2-amino-3-methyl-3H-imidazo[4,5-f]quinoline (IQ) and probiotic lactobacilli: Lb. casei 0900, Lb. casei 0908, and Lb. paracasei 0919. Our results show that the carcinogen IQ greatly increased FW genotoxicity (up to 16.92 ± 3.03 U/mg) and the activity of fecal enzymes (up to even 1.4 ± 0.16 U/mg) in 15 individuals (children, adults and elderly). After incubation with IQ, the activity of β-glucuronidase was reduced by Lactobacillus bacteria by 76.0% (Lb. paracasei 0908) in the FW of children, and by 82.0% (Lb. paracasei 0919) in the elderly; while that of β-glucosidase was reduced by 55.0% in children (Lb. casei 0908) and 90.0% (Lb. paracasei 0919) in elderly subjects. Lactobacilli decreased the genotoxicity of FW after incubation with IQ to the greatest extent in adults (by 64.5%). Probiotic lactobacilli, in the presence of IQ, efficiently inhibits activity of fecal enzymes to the level of control. Genotoxicity inhibition depends on the person's age, its individual microbiota and diet.
Collapse
Affiliation(s)
- Adriana Nowak
- Institute of Fermentation Technology and Microbiology, Department of Biotechnology and Food Sciences, Lodz University of Technology, Wolczanska 171/173, 90-924 Lodz, Poland.
| | - Katarzyna Śliżewska
- Institute of Fermentation Technology and Microbiology, Department of Biotechnology and Food Sciences, Lodz University of Technology, Wolczanska 171/173, 90-924 Lodz, Poland
| |
Collapse
|
129
|
High-resolution time-of-flight mass spectrometry fingerprinting of metabolites from cecum and distal colon contents of rats fed resistant starch. Anal Bioanal Chem 2013; 406:745-56. [DOI: 10.1007/s00216-013-7523-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Revised: 11/14/2013] [Accepted: 11/18/2013] [Indexed: 10/25/2022]
|
130
|
Ramakrishna BS. Role of the gut microbiota in human nutrition and metabolism. J Gastroenterol Hepatol 2013; 28 Suppl 4:9-17. [PMID: 24251697 DOI: 10.1111/jgh.12294] [Citation(s) in RCA: 298] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/14/2013] [Indexed: 02/06/2023]
Abstract
The human gastrointestinal tract harbors trillions of bacteria, most of which are commensal and have adapted over time to the milieu of the human colon. Their many metabolic interactions with each other, and with the human host, influence human nutrition and metabolism in diverse ways. Our understanding of these influences has come through breakthroughs in the molecular profiling of the phylogeny and the metabolic capacities of the microbiota. The gut microbiota produce a variety of nutrients including short-chain fatty acids, B vitamins, and vitamin K. Because of their ability to interact with receptors on epithelial cells and subepithelial cells, the microbiota also release a number of cellular factors that influence human metabolism. Thus, they have potential roles in the pathogenesis of metabolic syndrome, diabetes, non-alcoholic fatty liver disease, and cognition, which extend well beyond their traditional contribution to nutrition. This review explores the roles of the gut microbiota in human nutrition and metabolism, and the putative mechanisms underlying these effects.
Collapse
Affiliation(s)
- Balakrishnan S Ramakrishna
- Department of Gastroenterology, SRM Institutes of Medical Science, Chennai, India; Flinders University of South Australia, Bedford Park, South Australia, Australia
| |
Collapse
|
131
|
Kato I, Startup J, Ram JL. Fecal Biomarkers for Research on Dietary and Lifestyle Risk Factors in Colorectal Cancer Etiology. CURRENT COLORECTAL CANCER REPORTS 2013. [DOI: 10.1007/s11888-013-0195-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
132
|
Effects of dietary nutrients on volatile breath metabolites. J Nutr Sci 2013; 2:e34. [PMID: 25191584 PMCID: PMC4153095 DOI: 10.1017/jns.2013.26] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2013] [Revised: 07/22/2013] [Accepted: 07/25/2013] [Indexed: 12/22/2022] Open
Abstract
Breath analysis is becoming increasingly established as a means of assessing metabolic,
biochemical and physiological function in health and disease. The methods available for
these analyses exploit a variety of complex physicochemical principles, but are becoming
more easily utilised in the clinical setting. Whilst some of the factors accounting for
the biological variation in breath metabolite concentrations have been clarified, there
has been relatively little work on the dietary factors that may influence them. In
applying breath analysis to the clinical setting, it will be important to consider how
these factors may affect the interpretation of endogenous breath composition. Diet may
have complex effects on the generation of breath compounds. These effects may either be
due to a direct impact on metabolism, or because they alter the gastrointestinal flora.
Bacteria are a major source of compounds in breath, and their generation of H2,
hydrogen cyanide, aldehydes and alkanes may be an indicator of the health of their
host.
Collapse
|
133
|
An C, Kuda T, Yazaki T, Takahashi H, Kimura B. Caecal fermentation, putrefaction and microbiotas in rats fed milk casein, soy protein or fish meal. Appl Microbiol Biotechnol 2013; 98:2779-87. [PMID: 24113824 DOI: 10.1007/s00253-013-5271-5] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Revised: 09/13/2013] [Accepted: 09/16/2013] [Indexed: 02/07/2023]
Abstract
To clarify the effect of soy protein (SP) and fish meal (FM), compared to milk casein (MC), on the intestinal environment, we examined caecal environment of rats fed the test diets. Four-week-old rats were fed AIN-76-based diet containing 20 %, w/w MC, SP or FM for 16 days. Caecal organic acids were analysed by HPLC. Caecal putrefactive compounds (indole, phenol, H2S and ammonia) were analysed by colorimetric assays. Caecal microflora was determined by 16S rRNA gene-DGGE and pyrosequencing with bar-coded primers targeting the bacterial 16S rRNA gene. n-Butyric and lactic acid levels were high in rats fed SP and FM, respectively. Butyrate-producing bacteria, such as Oscillibacter, and lactate-producing bacteria, such as Lactobacillus, were detected in each diet group. Also, the putrefactive compound contents were high in rats fed SP and FM. In this study, both DGGE and pyrosequencing analyses were able to evaluate the dynamics of the intestinal microbiota. The results indicate that dietary proteins can alter the intestinal environment, affecting fermentation by the intestinal microbiota and the generation of putrefactive compounds.
Collapse
Affiliation(s)
- Choa An
- Department of Food Science and Technology, Tokyo University of Marine Science and Technology, 4-5-7 Konan, Minato-ku, Tokyo, Japan
| | | | | | | | | |
Collapse
|
134
|
Rossi M, Amaretti A, Leonardi A, Raimondi S, Simone M, Quartieri A. Potential impact of probiotic consumption on the bioactivity of dietary phytochemicals. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2013; 61:9551-9558. [PMID: 24007212 DOI: 10.1021/jf402722m] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Many healthy phytochemicals occur in food in the form of esters, glycoconjugates, or polymers, which are not directly bioavailable. Probiotic lactobacilli and bifidobacteria, which have evolved within the colonic ecosystem where indigestible oligo- and polysaccharides are their sole carbon sources, bear several glycosyl-hydrolases and can contribute to release the aglycones from glycoconjugated phytochemicals. Among the glycosyl-hydrolases, β-glucosidases are the most pertinent, because many phytochemicals are glucoconjugates. β-Glucosidase-positive probiotic bacteria were proved to release the aglycones of isoflavones and lignans in vitro, but studies in vivo are scarce. A positive correlation between probiotic consumption and urinary and/or plasma levels of isoflavone or lignan metabolites was not established. However, the strains used in the trials were not validated for the enzymatic properties or for the ability to hydrolyze lignans or isoflavones. Thus, activation of specific phytochemicals by probiotic bacteria still needs substantial efforts to be proved.
Collapse
Affiliation(s)
- Maddalena Rossi
- Department of Life Sciences, University of Modena and Reggio Emilia , 41125 Modena, Italy
| | | | | | | | | | | |
Collapse
|
135
|
Chong ESL. A potential role of probiotics in colorectal cancer prevention: review of possible mechanisms of action. World J Microbiol Biotechnol 2013; 30:351-74. [PMID: 24068536 DOI: 10.1007/s11274-013-1499-6] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Accepted: 09/16/2013] [Indexed: 02/07/2023]
Abstract
A number of investigations, mainly using in vitro and animal models, have demonstrated a wide range of possible mechanisms, by which probiotics may play a role in colorectal cancer (CRC) prevention. In this context, the most well studied probiotics are certain strains from the genera of lactobacilli and bifidobacteria. The reported anti-CRC mechanisms of probiotics encompass intraluminal, systemic, and direct effects on intestinal mucosa. Intraluminal effects detailed in this review include competitive exclusion of pathogenic intestinal flora, alteration of intestinal microflora enzyme activity, reduction of carcinogenic secondary bile acids, binding of carcinogens and mutagens, and increasing short chain fatty acids production. Reduction of DNA damage and suppression of aberrant crypt foci formation have been well demonstrated as direct anti-CRC effects of probiotics on intestinal mucosa. Existing evidence clearly support a multifaceted immunomodulatory role of probiotics in CRC, particularly its ability to modulate intestinal inflammation, a well known risk factor for CRC. The effectiveness of probiotics in CRC prevention is dependent on the strain of the microorganism, while viability may not be a prerequisite for certain probiotic anticancer mechanisms, as indicated by several studies. Emerging data suggest synbiotic as a more effective approach than either prebiotics or probiotics alone. More in vivo especially human studies are warranted to further elucidate and confirm the potential role of probiotics (viable and non-viable), prebiotics and synbiotics in CRC chemoprevention.
Collapse
Affiliation(s)
- Esther Swee Lan Chong
- Institute of Food, Nutrition and Human Health, Massey University, PO Box 11222, Palmerston North, 4442, New Zealand,
| |
Collapse
|
136
|
Abstract
BACKGROUND The aim of this study was to determine whether volatile organic compounds (VOCs) present in the headspace of feces could be used to diagnose or distinguish between chronic diseases of the gastrointestinal tract and apparently healthy volunteers. METHODS A total of 87 people were recruited, divided between 4 categories: healthy volunteers (n = 19), Crohn's disease (n = 22), ulcerative colitis (n = 20), and irritable bowel syndrome (n = 26). They each supplied fecal samples before, and except for the healthy volunteers, after treatment. Fecal samples were incubated in a sample bag with added purified air at 40°C and headspace samples were taken and concentrated on thermal sorption tubes. Gas chromatography-mass spectrometry then desorbed and analyzed these. The concentrations of a selection of high-abundance compounds were determined and assessed for differences in concentration between the groups. RESULTS Crohn's disease samples showed significant elevations in the concentrations of ester and alcohol derivates of short-chain fatty acids and indole compared with the other groups; indole and phenol were elevated in ulcerative colitis and irritable bowel syndrome but not at a statistically significant level. After treatment, the levels of many of the VOCs were significantly reduced and were more similar to those concentrations in healthy controls. CONCLUSIONS The abundance of a number of VOCs in feces differs markedly between Crohn's disease and other gastrointestinal conditions. Following treatment, the VOC profile is altered to more closely resemble that of healthy volunteers.
Collapse
|
137
|
Khalil NA, Walton GE, Gibson GR, Tuohy KM, Andrews SC. In vitro batch cultures of gut microbiota from healthy and ulcerative colitis (UC) subjects suggest that sulphate-reducing bacteria levels are raised in UC and by a protein-rich diet. Int J Food Sci Nutr 2013; 65:79-88. [PMID: 23941288 DOI: 10.3109/09637486.2013.825700] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Imbalances in gut microbiota composition during ulcerative colitis (UC) indicate a role for the microbiota in propagating the disorder. Such effects were investigated using in vitro batch cultures (with/without mucin, peptone or starch) inoculated with faecal slurries from healthy or UC patients; the growth of five bacterial groups was monitored along with short-chain fatty acid (SCFA) production. Healthy cultures gave two-fold higher growth and SCFA levels with up to ten-fold higher butyrate production. Starch gave the highest growth and SCFA production (particularly butyrate), indicating starch-enhanced saccharolytic activity. Sulphate-reducing bacteria (SRB) were the predominant bacterial group (of five examined) for UC inocula whereas they were the minority group for the healthy inocula. Furthermore, SRB growth was stimulated by peptone presumably due to the presence of sulphur-rich amino acids. The results suggest raised SRB levels in UC, which could contribute to the condition through release of toxic sulphide.
Collapse
|
138
|
Enos MK, Burton JP, Dols J, Buhulata S, Changalucha J, Reid G. Probiotics and nutrients for the first 1000 days of life in the developing world. Benef Microbes 2013; 4:3-16. [PMID: 23257014 DOI: 10.3920/bm2012.0020] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Clinically proven probiotics are, for the most part, not available in the developing world and certainly not affordable for the majority of people. This is unconscionable considering these products can alleviate diarrhoea and various infections, which are by far the major cause of death in children and in adults who are HIV positive. Indeed, some of these products have been proven in developing world settings. Distribution networks exist along with pharmacies and clinics that dispense drugs and products that require refrigeration. So, are lack of profit or company resources the problem? Our university has shown that alternative community based kitchen models that produce probiotics can be established. These empower local people, are socially responsible, produce affordable products and deliver benefits to over 3,000 children and adults daily. Surely, other institutions and corporations can multiply this effect and develop social business models across the developing world that are supported by clinical and basic science studies? In this review, we will discuss the application of probiotics and selected nutrients in the first 1000 days of life, a critical timepoint which is particularly challenging in resource disadvantaged countries.
Collapse
Affiliation(s)
- M K Enos
- Canadian Research and Development Centre for Probiotics, F2-116, The Lawson Health Research Institute, 268 Grosvenor Street, London, ON N6A 4V2, Canada
| | | | | | | | | | | |
Collapse
|
139
|
Kim MH, Kang SG, Park JH, Yanagisawa M, Kim CH. Short-chain fatty acids activate GPR41 and GPR43 on intestinal epithelial cells to promote inflammatory responses in mice. Gastroenterology 2013; 145:396-406.e1-10. [PMID: 23665276 DOI: 10.1053/j.gastro.2013.04.056] [Citation(s) in RCA: 686] [Impact Index Per Article: 62.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Revised: 04/04/2013] [Accepted: 04/25/2013] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS Short-chain fatty acids (SCFAs), the most abundant microbial metabolites in the intestine, activate cells via G-protein-coupled receptors (GPRs), such as GPR41 and GPR43. We studied regulation of the immune response by SCFAs and their receptors in the intestines of mice. METHODS Inflammatory responses were induced in GPR41(-/-), GPR43(-/-), and C57BL6 (control) mice by administration of ethanol; 2, 4, 6-trinitrobenzene sulfonic-acid (TNBS); or infection with Citrobacter rodentium. We examined the effects of C rodentium infection on control mice fed SCFAs and/or given injections of antibodies that delay the immune response. We also studied the kinetics of cytokine and chemokine production, leukocyte recruitment, intestinal permeability, and T-cell responses. Primary colon epithelial cells were isolated from GPR41(-/-), GPR43(-/-), and control mice; signaling pathways regulated by SCFAs were identified using immunohistochemical, enzyme-linked immunosorbent assay, and flow cytometry analyses. RESULTS GPR41(-/-) and GPR43(-/-) mice had reduced inflammatory responses after administration of ethanol or TNBS compared with control mice, and had a slower immune response against C rodentium infection, clearing the bacteria more slowly. SCFAs activated intestinal epithelial cells to produce chemokines and cytokines in culture and mice after administration of ethanol, TNBS, or C rodentium. These processes required GPR41 and GPR43 and were required to recruit leukocytes and activate effector T cells in the intestine. GPR41 and GPR43 activated extracellular signal-regulated kinase 1/2 and p38 mitogen-activated protein kinase signaling pathways in epithelial cells to induce production of chemokines and cytokines during immune responses. CONCLUSIONS SCFAs activate GPR41 and GPR43 on intestinal epithelial cells, leading to mitogen-activated protein kinase signaling and rapid production of chemokines and cytokines. These pathways mediate protective immunity and tissue inflammation in mice.
Collapse
Affiliation(s)
- Myung H Kim
- Laboratory of Immunology and Hematopoiesis, Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana 47907, USA
| | | | | | | | | |
Collapse
|
140
|
Shen X, Carlström M, Borniquel S, Jädert C, Kevil CG, Lundberg J. Microbial regulation of host hydrogen sulfide bioavailability and metabolism. Free Radic Biol Med 2013; 60:195-200. [PMID: 23466556 PMCID: PMC4077044 DOI: 10.1016/j.freeradbiomed.2013.02.024] [Citation(s) in RCA: 134] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Accepted: 02/22/2013] [Indexed: 10/27/2022]
Abstract
Hydrogen sulfide (H2S), generated through various endogenous enzymatic and nonenzymatic pathways, is emerging as a regulator of physiological and pathological events throughout the body. Bacteria in the gastrointestinal tract also produce significant amounts of H2S that regulates microflora growth and virulence responses. However, the impact of the microbiota on host global H2S bioavailability and metabolism remains unknown. To address this question, we examined H2S bioavailability in its various forms (free, acid labile, or bound sulfane sulfur), cystathionine γ-lyase (CSE) activity, and cysteine levels in tissues from germ-free versus conventionally housed mice. Free H2S levels were significantly reduced in plasma and gastrointestinal tissues of germ-free mice. Bound sulfane sulfur levels were decreased by 50-80% in germ-free mouse plasma and adipose and lung tissues. Tissue CSE activity was significantly reduced in many organs from germ-free mice, whereas tissue cysteine levels were significantly elevated compared to conventional mice. These data reveal that the microbiota profoundly regulates systemic bioavailability and metabolism of H2S.
Collapse
Affiliation(s)
- Xinggui Shen
- Department of Pathology, Molecular and Cellular Physiology and Cell Biology and Anatomy, LSU Health-Shreveport
| | - Mattias Carlström
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Sara Borniquel
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Cecilia Jädert
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Christopher G Kevil
- Department of Pathology, Molecular and Cellular Physiology and Cell Biology and Anatomy, LSU Health-Shreveport
- Correspondence to: Christopher Kevil, PhD, LSU Health-Shreveport, or Jon Lundberg, MD, PhD, Karolinska Institutet,
| | - Jon Lundberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- Correspondence to: Christopher Kevil, PhD, LSU Health-Shreveport, or Jon Lundberg, MD, PhD, Karolinska Institutet,
| |
Collapse
|
141
|
Stecher B, Berry D, Loy A. Colonization resistance and microbial ecophysiology: using gnotobiotic mouse models and single-cell technology to explore the intestinal jungle. FEMS Microbiol Rev 2013; 37:793-829. [PMID: 23662775 DOI: 10.1111/1574-6976.12024] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Revised: 04/11/2013] [Accepted: 04/18/2013] [Indexed: 12/14/2022] Open
Abstract
The highly diverse intestinal microbiota forms a structured community engaged in constant communication with itself and its host and is characterized by extensive ecological interactions. A key benefit that the microbiota affords its host is its ability to protect against infections in a process termed colonization resistance (CR), which remains insufficiently understood. In this review, we connect basic concepts of CR with new insights from recent years and highlight key technological advances in the field of microbial ecology. We present a selection of statistical and bioinformatics tools used to generate hypotheses about synergistic and antagonistic interactions in microbial ecosystems from metagenomic datasets. We emphasize the importance of experimentally testing these hypotheses and discuss the value of gnotobiotic mouse models for investigating specific aspects related to microbiota-host-pathogen interactions in a well-defined experimental system. We further introduce new developments in the area of single-cell analysis using fluorescence in situ hybridization in combination with metabolic stable isotope labeling technologies for studying the in vivo activities of complex community members. These approaches promise to yield novel insights into the mechanisms of CR and intestinal ecophysiology in general, and give researchers the means to experimentally test hypotheses in vivo at varying levels of biological and ecological complexity.
Collapse
Affiliation(s)
- Bärbel Stecher
- Max von Pettenkofer Institute of Hygiene and Medical Microbiology, Ludwig-Maximilians-University of Munich, Munich, Germany.
| | | | | |
Collapse
|
142
|
McHardy IH, Goudarzi M, Tong M, Ruegger PM, Schwager E, Weger JR, Graeber TG, Sonnenburg JL, Horvath S, Huttenhower C, McGovern DPB, Fornace AJ, Borneman J, Braun J. Integrative analysis of the microbiome and metabolome of the human intestinal mucosal surface reveals exquisite inter-relationships. MICROBIOME 2013; 1:17. [PMID: 24450808 PMCID: PMC3971612 DOI: 10.1186/2049-2618-1-17] [Citation(s) in RCA: 204] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2013] [Accepted: 05/12/2013] [Indexed: 05/10/2023]
Abstract
BACKGROUND Consistent compositional shifts in the gut microbiota are observed in IBD and other chronic intestinal disorders and may contribute to pathogenesis. The identities of microbial biomolecular mechanisms and metabolic products responsible for disease phenotypes remain to be determined, as do the means by which such microbial functions may be therapeutically modified. RESULTS The composition of the microbiota and metabolites in gut microbiome samples in 47 subjects were determined. Samples were obtained by endoscopic mucosal lavage from the cecum and sigmoid colon regions, and each sample was sequenced using the 16S rRNA gene V4 region (Illumina-HiSeq 2000 platform) and assessed by UPLC mass spectroscopy. Spearman correlations were used to identify widespread, statistically significant microbial-metabolite relationships. Metagenomes for identified microbial OTUs were imputed using PICRUSt, and KEGG metabolic pathway modules for imputed genes were assigned using HUMAnN. The resulting metabolic pathway abundances were mostly concordant with metabolite data. Analysis of the metabolome-driven distribution of OTU phylogeny and function revealed clusters of clades that were both metabolically and metagenomically similar. CONCLUSIONS The results suggest that microbes are syntropic with mucosal metabolome composition and therefore may be the source of and/or dependent upon gut epithelial metabolites. The consistent relationship between inferred metagenomic function and assayed metabolites suggests that metagenomic composition is predictive to a reasonable degree of microbial community metabolite pools. The finding that certain metabolites strongly correlate with microbial community structure raises the possibility of targeting metabolites for monitoring and/or therapeutically manipulating microbial community function in IBD and other chronic diseases.
Collapse
Affiliation(s)
- Ian H McHardy
- Pathology and Laboratory Medicine UCLA, Los Angeles, CA, USA
| | - Maryam Goudarzi
- Biochemistry and Molecular and Cellular Biology, Georgetown University, Washington, DC, USA
| | - Maomeng Tong
- Molecular and Medical Pharmacology, UCLA, Los Angeles, CA, USA
| | | | | | - John R Weger
- Plant Pathology, UC Riverside, Riverside, CA, USA
| | | | | | | | | | - Dermot PB McGovern
- The F. Widjaja Family Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedar's Sinai Medical Center, Los Angeles, CA, USA
| | - Albert J Fornace
- Biochemistry and Molecular and Cellular Biology, Georgetown University, Washington, DC, USA
| | | | - Jonathan Braun
- Pathology and Laboratory Medicine UCLA, Los Angeles, CA, USA
| |
Collapse
|
143
|
Modulation of the fecal bile acid profile by gut microbiota in cirrhosis. J Hepatol 2013; 58:949-55. [PMID: 23333527 PMCID: PMC3936319 DOI: 10.1016/j.jhep.2013.01.003] [Citation(s) in RCA: 567] [Impact Index Per Article: 51.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2012] [Revised: 11/01/2012] [Accepted: 01/03/2013] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS The 7α-dehydroxylation of primary bile acids (BAs), chenodeoxycholic (CDCA) and cholic acid (CA) into the secondary BAs, lithocholic (LCA) and deoxycholic acid (DCA), is a key function of the gut microbiota. We aimed at studying the linkage between fecal BAs and gut microbiota in cirrhosis since this could help understand cirrhosis progression. METHODS Fecal microbiota were analyzed by culture-independent multitagged-pyrosequencing, fecal BAs using HPLC and serum BAs using LC-MS in controls, early (Child A) and advanced cirrhotics (Child B/C). A subgroup of early cirrhotics underwent BA and microbiota analysis before/after eight weeks of rifaximin. RESULTS Cross-sectional: 47 cirrhotics (24 advanced) and 14 controls were included. In feces, advanced cirrhotics had the lowest total, secondary, secondary/primary BA ratios, and the highest primary BAs compared to early cirrhotics and controls. Secondary fecal BAs were detectable in all controls but in a significantly lower proportion of cirrhotics (p<0.002). Serum primary BAs were higher in advanced cirrhotics compared to the rest. Cirrhotics, compared to controls, had a higher Enterobacteriaceae (potentially pathogenic) but lower Lachonospiraceae, Ruminococcaceae and Blautia (7α-dehydroxylating bacteria) abundance. CDCA was positively correlated with Enterobacteriaceae (r=0.57, p<0.008) while Ruminococcaceae were positively correlated with DCA (r=0.4, p<0.05). A positive correlation between Ruminococcaceae and DCA/CA (r=0.82, p<0.012) and Blautia with LCA/CDCA (r=0.61, p<0.03) was also seen. Prospective study: post-rifaximin, six early cirrhotics had reduction in Veillonellaceae and in secondary/primary BA ratios. CONCLUSIONS Cirrhosis, especially advanced disease, is associated with a decreased conversion of primary to secondary fecal BAs, which is linked to abundance of key gut microbiome taxa.
Collapse
|
144
|
Altered gut microbiota promotes colitis-associated cancer in IL-1 receptor-associated kinase M-deficient mice. Inflamm Bowel Dis 2013; 19:1266-77. [PMID: 23567778 PMCID: PMC3744230 DOI: 10.1097/mib.0b013e318281330a] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Microbial sensing by Toll-like receptors (TLR) and its negative regulation have an important role in the pathogenesis of inflammation-related cancer. In this study, we investigated the role of negative regulation of Toll-like receptors signaling and gut microbiota in the development of colitis-associated cancer in mouse model. METHODS Colitis-associated cancer was induced by azoxymethane and dextran sodium sulfate in wild-type and in interleukin-1 receptor-associated kinase M (IRAK-M)-deficient mice with or without antibiotic (ATB) treatment. Local cytokine production was analyzed by multiplex cytokine assay or enzyme-linked immunosorbent assay, and regulatory T cells were analyzed by flow cytometry. Changes in microbiota composition during tumorigenesis were analyzed by pyrosequencing, and β-glucuronidase activity was measured in intestinal content by fluorescence assay. RESULTS ATB treatment of wild-type mice reduced the incidence and severity of tumors. Compared with nontreated mice, ATB-treated mice had significantly lower numbers of regulatory T cells in colon, altered gut microbiota composition, and decreased β-glucuronidase activity. However, the β-glucuronidase activity was not as low as in germ-free mice. IRAK-M-deficient mice not only developed invasive tumors, but ATB-induced decrease in β-glucuronidase activity did not rescue them from severe carcinogenesis phenotype. Furthermore, IRAK-M-deficient mice had significantly increased levels of proinflammatory cytokines in the tumor tissue. CONCLUSIONS We conclude that gut microbiota promotes tumorigenesis by increasing the exposure of gut epithelium to carcinogens and that IRAK-M-negative regulation is essential for colon cancer resistance even in conditions of altered microbiota. Therefore, gut microbiota and its metabolic activity could be potential targets for colitis-associated cancer therapy.
Collapse
|
145
|
Bajaj JS, Heuman DM, Sanyal AJ, Hylemon PB, Sterling RK, Stravitz RT, Fuchs M, Ridlon JM, Daita K, Monteith P, Noble NA, White MB, Fisher A, Sikaroodi M, Rangwala H, Gillevet PM. Modulation of the metabiome by rifaximin in patients with cirrhosis and minimal hepatic encephalopathy. PLoS One 2013; 8:e60042. [PMID: 23565181 PMCID: PMC3615021 DOI: 10.1371/journal.pone.0060042] [Citation(s) in RCA: 314] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Accepted: 02/19/2013] [Indexed: 12/12/2022] Open
Abstract
UNLABELLED Hepatic encephalopathy (HE) represents a dysfunctional gut-liver-brain axis in cirrhosis which can negatively impact outcomes. This altered gut-brain relationship has been treated using gut-selective antibiotics such as rifaximin, that improve cognitive function in HE, especially its subclinical form, minimal HE (MHE). However, the precise mechanism of the action of rifaximin in MHE is unclear. We hypothesized that modulation of gut microbiota and their end-products by rifaximin would affect the gut-brain axis and improve cognitive performance in cirrhosis. Aim To perform a systems biology analysis of the microbiome, metabolome and cognitive change after rifaximin in MHE. METHODS Twenty cirrhotics with MHE underwent cognitive testing, endotoxin analysis, urine/serum metabolomics (GC and LC-MS) and fecal microbiome assessment (multi-tagged pyrosequencing) at baseline and 8 weeks post-rifaximin 550 mg BID. Changes in cognition, endotoxin, serum/urine metabolites (and microbiome were analyzed using recommended systems biology techniques. Specifically, correlation networks between microbiota and metabolome were analyzed before and after rifaximin. RESULTS There was a significant improvement in cognition(six of seven tests improved, p<0.01) and endotoxemia (0.55 to 0.48 Eu/ml, p = 0.02) after rifaximin. There was a significant increase in serum saturated (myristic, caprylic, palmitic, palmitoleic, oleic and eicosanoic) and unsaturated (linoleic, linolenic, gamma-linolenic and arachnidonic) fatty acids post-rifaximin. No significant microbial change apart from a modest decrease in Veillonellaceae and increase in Eubacteriaceae was observed. Rifaximin resulted in a significant reduction in network connectivity and clustering on the correlation networks. The networks centered on Enterobacteriaceae, Porphyromonadaceae and Bacteroidaceae indicated a shift from pathogenic to beneficial metabolite linkages and better cognition while those centered on autochthonous taxa remained similar. CONCLUSIONS Rifaximin is associated with improved cognitive function and endotoxemia in MHE, which is accompanied by alteration of gut bacterial linkages with metabolites without significant change in microbial abundance. TRIAL REGISTRATION ClinicalTrials.gov NCT01069133.
Collapse
Affiliation(s)
- Jasmohan S Bajaj
- Division of Gastroenterology, Hepatology and Nutrition, Virginia Commonwealth University and McGuire Veterans Affairs Medical Center, Richmond, Virginia, United States of America.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
146
|
Biagi E, Candela M, Turroni S, Garagnani P, Franceschi C, Brigidi P. Ageing and gut microbes: perspectives for health maintenance and longevity. Pharmacol Res 2013; 69:11-20. [PMID: 23079287 DOI: 10.1016/j.phrs.2012.10.005] [Citation(s) in RCA: 160] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2012] [Revised: 10/01/2012] [Accepted: 10/06/2012] [Indexed: 12/13/2022]
Abstract
The ageing process affects the human gut microbiota phylogenetic composition and its interaction with the immune system. Age-related gut microbiota modifications are associated with immunosenescence and inflamm-ageing in a sort of self-sustaining loop, which allows the placement of gut microbiota unbalances among both the causes and the effects of the inflamm-ageing process. Even if, up to now, the link between gut microbiota and the ageing process is only partially understood, the gut ecosystem shows the potential to become a promising target for strategies able to contribute to the health status of older people. In this context, the consumption of pro/prebiotics may be useful in both prevention and treatment of age-related pathophysiological conditions, such as recovery and promotion of immune functions, i.e. adjuvant effect for influenza vaccine, and prevention and/or alleviation of common "winter diseases", as well as constipation and Clostridium difficile-associated diarrhoea. Moreover, being involved in different mechanisms which concur in counteracting inflammation, such as down-regulation of inflammation-associated genes and improvement of colonic mucosa conditions, probiotics have the potentiality to be involved in the promotion of longevity.
Collapse
Affiliation(s)
- Elena Biagi
- Department of Pharmaceutical Sciences, University of Bologna, Via Belmeloro 6, 40126, Bologna, Italy
| | | | | | | | | | | |
Collapse
|
147
|
Davila AM, Blachier F, Gotteland M, Andriamihaja M, Benetti PH, Sanz Y, Tomé D. Intestinal luminal nitrogen metabolism: Role of the gut microbiota and consequences for the host. Pharmacol Res 2013. [DOI: 10.1016/j.phrs.2012.11.005] [Citation(s) in RCA: 248] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
148
|
Davila AM, Blachier F, Gotteland M, Andriamihaja M, Benetti PH, Sanz Y, Tomé D. Re-print of "Intestinal luminal nitrogen metabolism: role of the gut microbiota and consequences for the host". Pharmacol Res 2013; 69:114-26. [PMID: 23318949 DOI: 10.1016/j.phrs.2013.01.003] [Citation(s) in RCA: 139] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Alimentary and endogenous proteins are mixed in the small intestinal lumen with the microbiota. Although experimental evidences suggest that the intestinal microbiota is able to incorporate and degrade some of the available amino acids, it appears that the microbiota is also able to synthesize amino acids raising the view that amino acid exchange between the microbiota and host can proceed in both directions. Although the net result of such exchanges remains to be determined, it is likely that a significant part of the amino acids recovered from the alimentary proteins are used by the microbiota. In the large intestine, where the density of bacteria is much higher than in the small intestine and the transit time much longer, the residual undigested luminal proteins and peptides can be degraded in amino acids by the microbiota. These amino acids cannot be absorbed to a significant extent by the colonic epithelium, but are precursors for the synthesis of numerous metabolic end products in reactions made by the microbiota. Among these products, some like short-chain fatty acids and organic acids are energy substrates for the colonic mucosa and several peripheral tissues while others like sulfide and ammonia can affect the energy metabolism of colonic epithelial cells. More work is needed to clarify the overall effects of the intestinal microbiota on nitrogenous compound metabolism and consequences on gut and more generally host health.
Collapse
Affiliation(s)
- Anne-Marie Davila
- UMR 914 INRA/AgroParisTech, Nutrition Physiology and Ingestive Behavior, Paris, France.
| | | | | | | | | | | | | |
Collapse
|
149
|
Scott KP, Gratz SW, Sheridan PO, Flint HJ, Duncan SH. The influence of diet on the gut microbiota. Pharmacol Res 2012; 69:52-60. [PMID: 23147033 DOI: 10.1016/j.phrs.2012.10.020] [Citation(s) in RCA: 640] [Impact Index Per Article: 53.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Revised: 10/15/2012] [Accepted: 10/29/2012] [Indexed: 02/06/2023]
Abstract
Diet is a major factor driving the composition and metabolism of the colonic microbiota. The amount, type and balance of the main dietary macronutrients (carbohydrates, proteins and fats) have a great impact on the large intestinal microbiota. The human colon contains a dense population of bacterial cells that outnumber host cells 10-fold. Bacteroidetes, Firmicutes and Actinobacteria are the three major phyla that inhabit the human large intestine and these bacteria possess a fascinating array of enzymes that can degrade complex dietary substrates. Certain colonic bacteria are able to metabolise a remarkable variety of substrates whilst other species carry out more specialised activities, including primary degradation of plant cell walls. Microbial metabolism of dietary carbohydrates results mainly in the formation of short chain fatty acids and gases. The major bacterial fermentation products are acetate, propionate and butyrate; and the production of these tends to lower the colonic pH. These weak acids influence the microbial composition and directly affect host health, with butyrate the preferred energy source for the colonocytes. Certain bacterial species in the colon survive by cross-feeding, using either the breakdown products of complex carbohydrate degradation or fermentation products such as lactic acid for growth. Microbial protein metabolism results in additional fermentation products, some of which are potentially harmful to host health. The current 'omic era promises rapid progress towards understanding how diet can be used to modulate the composition and metabolism of the gut microbiota, allowing researchers to provide informed advice, that should improve long-term health status.
Collapse
Affiliation(s)
- Karen P Scott
- Microbiology Group, Rowett Institute of Nutrition and Health, University of Aberdeen, Greenburn Road, Bucksburn, Aberdeen AB21 9SB, UK.
| | | | | | | | | |
Collapse
|
150
|
Usami M, Miyoshi M, Kanbara Y, Aoyama M, Sakaki H, Shuno K, Hirata K, Takahashi M, Ueno K, Hamada Y, Tabata S, Asahara T, Nomoto K. Analysis of fecal microbiota, organic acids and plasma lipids in hepatic cancer patients with or without liver cirrhosis. Clin Nutr 2012; 32:444-51. [PMID: 23068014 DOI: 10.1016/j.clnu.2012.09.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2011] [Revised: 09/19/2012] [Accepted: 09/24/2012] [Indexed: 12/15/2022]
Abstract
BACKGROUND & AIMS Changes in the microbiota composition are able to affect nutrient absorption and energy metabolism, but there are few human studies. The aims were to analyze fecal constituents quantitatively and compare them with liver dysfunction in hepatic cancer patients and to evaluate the relationships among intestinal microbiota, fecal organic acids and plasma lipid composition. METHODS Fecal samples collected from 46 hepatic cancer patients (with liver cirrhosis, chronic hepatitis or liver fibrosis and normal liver) were evaluated for fecal constituents. Blood organic acid, lipid and fatty acid concentrations were analyzed. RESULTS Fecal microbiota and organic acids showed no significant differences among different liver dysfunction patients. In normal liver patients, fecal Candida was positively correlated with plasma phospholipid while Bifidobacterium was negatively correlated with plasma eicosapentaenoic acid and eicosapentaenoic acid/arachidonic acid ratio (all p < 0.05). In cirrhotic liver patients, positive correlations were noted for Lactobacillus and docosahexaenoic acid and Candida and eicosapentaenoic acid or eicosapentaenoic acid/arachidonic acid ratio (all p < 0.01). It was suggested that intestinal biota affected serum fatty acid metabolism and were modified by liver disorders. CONCLUSIONS Intestinal microbiota and organic acid concentrations in hepatic cancer patients had positive and/or negative correlations with serum lipid levels.
Collapse
Affiliation(s)
- Makoto Usami
- Division of Nutrition and Metabolism, Kobe University Graduate School of Health Sciences, Kobe, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|