3151
|
The effects of fermented wheat powder (Lisosan G) on the blood lipids and oxidative status of healthy rabbits. Food Chem Toxicol 2015; 84:1-7. [DOI: 10.1016/j.fct.2015.07.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2015] [Revised: 06/26/2015] [Accepted: 07/06/2015] [Indexed: 12/31/2022]
|
3152
|
Faghfoori Z, Pourghassem Gargari B, Saber Gharamaleki A, Bagherpour H, Yari Khosroushahi A. Cellular and molecular mechanisms of probiotics effects on colorectal cancer. J Funct Foods 2015; 18:463-472. [DOI: 10.1016/j.jff.2015.08.013] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
3153
|
Kumar A, Alrefai WA, Borthakur A, Dudeja PK. Lactobacillus acidophilus counteracts enteropathogenic E. coli-induced inhibition of butyrate uptake in intestinal epithelial cells. Am J Physiol Gastrointest Liver Physiol 2015; 309:G602-7. [PMID: 26272259 PMCID: PMC4593819 DOI: 10.1152/ajpgi.00186.2015] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 08/10/2015] [Indexed: 01/31/2023]
Abstract
Butyrate, a key short-chain fatty acid metabolite of colonic luminal bacterial action on dietary fiber, serves as a primary fuel for the colonocytes, ameliorates mucosal inflammation, and stimulates NaCl absorption. Absorption of butyrate into the colonocytes is essential for these intracellular effects. Monocarboxylate transporter 1 (MCT1) plays a major role in colonic luminal butyrate absorption. Previous studies (Tan J, McKenzie C, Potamitis M, Thorburn AN, Mackay CR, Macia L. Adv Immunol 121: 91-119, 2014.) showed decreased MCT1 expression and function in intestinal inflammation. We have previously shown (Borthakur A, Gill RK, Hodges K, Ramaswamy K, Hecht G, Dudeja PK. Am J Physiol Gastrointest Liver Physiol 290: G30-G35, 2006.) impaired butyrate absorption in human intestinal epithelial Caco-2 cells due to decreased MCT1 level at the apical cell surface following enteropathogenic E. coli (EPEC) infection. Current studies, therefore, examined the potential role of probiotic Lactobacilli in stimulating MCT1-mediated butyrate uptake and counteracting EPEC inhibition of MCT1 function. Of the five species of Lactobacilli, short-term (3 h) treatment with L. acidophilus (LA) significantly increased MCT1-mediated butyrate uptake in Caco-2 cells. Heat-killed LA was ineffective, whereas the conditioned culture supernatant of LA (LA-CS) was equally effective in stimulating MCT1 function, indicating that the effects are mediated by LA-secreted soluble factor(s). Furthermore, LA-CS increased apical membrane levels of MCT1 protein via decreasing its basal endocytosis, suggesting that LA-CS stimulation of butyrate uptake could be secondary to increased levels of MCT1 on the apical cell surface. LA-CS also attenuated EPEC inhibition of butyrate uptake and EPEC-mediated endocytosis of MCT1. Our studies highlight distinct role of specific LA-secreted molecules in modulating colonic butyrate absorption.
Collapse
Affiliation(s)
- Anoop Kumar
- 1Department of Medicine, Division of Gastroenterology & Hepatology, University of Illinois at Chicago, Chicago, Illinois; and
| | - Waddah A. Alrefai
- 1Department of Medicine, Division of Gastroenterology & Hepatology, University of Illinois at Chicago, Chicago, Illinois; and ,2Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois
| | - Alip Borthakur
- Department of Medicine, Division of Gastroenterology & Hepatology, University of Illinois at Chicago, Chicago, Illinois; and
| | - Pradeep K. Dudeja
- 1Department of Medicine, Division of Gastroenterology & Hepatology, University of Illinois at Chicago, Chicago, Illinois; and ,2Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois
| |
Collapse
|
3154
|
Majnik AV, Lane RH. The relationship between early-life environment, the epigenome and the microbiota. Epigenomics 2015; 7:1173-84. [DOI: 10.2217/epi.15.74] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
3155
|
Leung K, Thuret S. Gut Microbiota: A Modulator of Brain Plasticity and Cognitive Function in Ageing. Healthcare (Basel) 2015; 3:898-916. [PMID: 27417803 PMCID: PMC4934620 DOI: 10.3390/healthcare3040898] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 09/15/2015] [Accepted: 09/24/2015] [Indexed: 02/06/2023] Open
Abstract
Gut microbiota have recently been a topic of great interest in the field of microbiology, particularly their role in normal physiology and its influence on human health in disease. A large body of research has supported the presence of a pathway of communication between the gut and the brain, modulated by gut microbiota, giving rise to the term “microbiota-gut-brain” axis. It is now thought that, through this pathway, microbiota can affect behaviour and modulate brain plasticity and cognitive function in ageing. This review summarizes the evidence supporting the existence of such a connection and possible mechanisms of action whereby microbiota can influence the function of the central nervous system. Since normalisation of gut flora has been shown to prevent changes in behaviour, we further postulate on possible therapeutic targets to intervene with cognitive decline in ageing. The research poses various limitations, for example uncertainty about how this data translates to broad human populations. Nonetheless, the microbiota-gut-brain axis is an exciting field worthy of further investigation, particularly with regards to its implications on the ageing population.
Collapse
Affiliation(s)
- Katherine Leung
- Institute of Psychiatry, King's College London, The James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK.
| | - Sandrine Thuret
- Institute of Psychiatry, King's College London, The James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK.
| |
Collapse
|
3156
|
Unno T, Hisada T, Takahashi S. Hesperetin Modifies the Composition of Fecal Microbiota and Increases Cecal Levels of Short-Chain Fatty Acids in Rats. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2015; 63:7952-7957. [PMID: 26306898 DOI: 10.1021/acs.jafc.5b02649] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
There has been particular interest in the prebiotic-like effects of commonly consumed polyphenols. This study aimed to evaluate the effects of hesperidin (HD) and its aglycone hesperetin (HT), major flavonoids in citrus fruits, on the structure and activity of gut microbiota in rats. Rats ingested an assigned diet (a control diet, a 0.5% HT diet, or a 1.0% HD diet) for 3 weeks. Terminal restriction fragment length polymorphism analysis revealed that the proportion of Clostridium subcluster XIVa in the feces collected at the third week of feeding was significantly reduced by the HT diet: 19.8 ± 4.3% for the control diet versus 5.3 ± 1.5% for the HT diet (P < 0.01). There was a significant difference in the cecal pool of short-chain fatty acids (SCFA), the sum of acetic, propionic, and butyric acids, between the control diet (212 ± 71 μmol) and the HT diet (310 ± 51 μmol) (P < 0.05), whereas the HD diet exhibited no effects (245 ± 51 μmol). Interestingly, dietary HT resulted in a significant increase in the excretion of starch in the feces. HT, but not HD, might reduce starch digestion, and parts of undigested starch were utilized to produce SCFA by microbial fermentation in the large intestine.
Collapse
Affiliation(s)
- Tomonori Unno
- Department of Health and Nutrition, Tokyo Kasei Gakuin University , 22 Sanban-cho, Chiyoda-ku, Tokyo 102-8341, Japan
| | - Takayoshi Hisada
- TechnoSuruga Laboratory Company, Ltd., 330 Nagasaki, Shimizu-ku, Shizuoka 424-0065, Japan
| | - Shunsuke Takahashi
- TechnoSuruga Laboratory Company, Ltd., 330 Nagasaki, Shimizu-ku, Shizuoka 424-0065, Japan
| |
Collapse
|
3157
|
van der Beek CM, Bloemen JG, van den Broek MA, Lenaerts K, Venema K, Buurman WA, Dejong CH. Hepatic Uptake of Rectally Administered Butyrate Prevents an Increase in Systemic Butyrate Concentrations in Humans. J Nutr 2015; 145:2019-24. [PMID: 26156796 DOI: 10.3945/jn.115.211193] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 06/15/2015] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Short-chain fatty acids (SCFAs), fermentation products of undigested fibers, are considered beneficial for colonic health. High plasma concentrations are potentially harmful; therefore, information about systemic SCFA clearance is needed before therapeutic use of prebiotics or colonic SCFA administration. OBJECTIVE The aim of this study was to investigate the effect of rectal butyrate administration on SCFA interorgan exchange. METHODS Twelve patients (7 men; age: 66.4 ± 2.0 y; BMI 24.5 ± 1.4 kg/m(2)) undergoing upper abdominal surgery participated in this randomized placebo-controlled trial. During surgery, 1 group received a butyrate enema (100 mmol sodium butyrate/L; 60 mL; n = 7), and the other group a placebo (140 mmol 0.9% NaCl/L; 60 mL; n = 5). Before and 5, 15, and 30 min after administration, blood samples were taken from the radial artery, hepatic vein, and portal vein. Plasma SCFA concentrations were analyzed, and fluxes from portal-drained viscera, liver, and splanchnic area were calculated and used for the calculation of the incremental area under the curve (iAUC) over a 30-min period. RESULTS Rectal butyrate administration led to higher portal butyrate concentrations at 5 min compared with placebo (92.2 ± 27.0 μmol/L vs. 14.3 ± 3.4 μmol/L, respectively; P < 0.01). In the butyrate-treated group, iAUCs of gut release (282.8 ± 133.8 μmol/kg BW · 0.5 h) and liver uptake (-293.7 ± 136.0 μmol/kg BW · 0.5 h) of butyrate were greater than in the placebo group [-16.6 ± 13.4 μmol/kg BW · 0.5 h (gut release) and 16.0 ± 13.8 μmol/kg BW · 0.5 h (liver uptake); P = 0.01 and P < 0.05, respectively]. As a result, splanchnic butyrate release did not differ between groups. CONCLUSION After colonic butyrate administration, splanchnic butyrate release was prevented in patients undergoing upper abdominal surgery. These observations imply that therapeutic colonic SCFA administration at this dose is safe. The trial was registered at clinicaltrials.gov as NCT02271802.
Collapse
Affiliation(s)
- Christina M van der Beek
- Department of Surgery, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, Netherlands; Top Institute Food & Nutrition, Wageningen, Netherlands; and
| | - Johanne G Bloemen
- Department of Surgery, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, Netherlands; Top Institute Food & Nutrition, Wageningen, Netherlands; and
| | - Maartje A van den Broek
- Department of Surgery, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Kaatje Lenaerts
- Department of Surgery, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, Netherlands; Top Institute Food & Nutrition, Wageningen, Netherlands; and
| | - Koen Venema
- Top Institute Food & Nutrition, Wageningen, Netherlands; and Beneficial Microbes Consultancy, Wageningen, Netherlands
| | - Wim A Buurman
- Department of Surgery, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Cornelis H Dejong
- Department of Surgery, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, Netherlands; Top Institute Food & Nutrition, Wageningen, Netherlands; and
| |
Collapse
|
3158
|
Kraler M, Schedle K, Schwarz C, Domig KJ, Pichler M, Oppeneder A, Wetscherek W, Prückler M, Pignitter M, Pirker KF, Somoza V, Heine D, Kneifel W. Fermented and extruded wheat bran in piglet diets: impact on performance, intestinal morphology, microbial metabolites in chyme and blood lipid radicals. Arch Anim Nutr 2015; 69:378-98. [DOI: 10.1080/1745039x.2015.1075671] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
3159
|
den Besten G, Gerding A, van Dijk TH, Ciapaite J, Bleeker A, van Eunen K, Havinga R, Groen AK, Reijngoud DJ, Bakker BM. Protection against the Metabolic Syndrome by Guar Gum-Derived Short-Chain Fatty Acids Depends on Peroxisome Proliferator-Activated Receptor γ and Glucagon-Like Peptide-1. PLoS One 2015; 10:e0136364. [PMID: 26292284 PMCID: PMC4546369 DOI: 10.1371/journal.pone.0136364] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 07/31/2015] [Indexed: 12/20/2022] Open
Abstract
The dietary fiber guar gum has beneficial effects on obesity, hyperglycemia and hypercholesterolemia in both humans and rodents. The major products of colonic fermentation of dietary fiber, the short-chain fatty acids (SCFAs), have been suggested to play an important role. Recently, we showed that SCFAs protect against the metabolic syndrome via a signaling cascade that involves peroxisome proliferator-activated receptor (PPAR) γ repression and AMP-activated protein kinase (AMPK) activation. In this study we investigated the molecular mechanism via which the dietary fiber guar gum protects against the metabolic syndrome. C57Bl/6J mice were fed a high-fat diet supplemented with 0% or 10% of the fiber guar gum for 12 weeks and effects on lipid and glucose metabolism were studied. We demonstrate that, like SCFAs, also guar gum protects against high-fat diet-induced metabolic abnormalities by PPARγ repression, subsequently increasing mitochondrial uncoupling protein 2 expression and AMP/ATP ratio, leading to the activation of AMPK and culminating in enhanced oxidative metabolism in both liver and adipose tissue. Moreover, guar gum markedly increased peripheral glucose clearance, possibly mediated by the SCFA-induced colonic hormone glucagon-like peptide-1. Overall, this study provides novel molecular insights into the beneficial effects of guar gum on the metabolic syndrome and strengthens the potential role of guar gum as a dietary-fiber intervention.
Collapse
Affiliation(s)
- Gijs den Besten
- Center for Liver, Digestive and Metabolic Diseases, Department of Pediatrics & Systems Biology Center for Energy Metabolism and Ageing, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Netherlands Consortium for Systems Biology, Amsterdam, The Netherlands
| | - Albert Gerding
- Center for Liver, Digestive and Metabolic Diseases, Department of Pediatrics & Systems Biology Center for Energy Metabolism and Ageing, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Theo H. van Dijk
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jolita Ciapaite
- Center for Liver, Digestive and Metabolic Diseases, Department of Pediatrics & Systems Biology Center for Energy Metabolism and Ageing, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Aycha Bleeker
- Center for Liver, Digestive and Metabolic Diseases, Department of Pediatrics & Systems Biology Center for Energy Metabolism and Ageing, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Top Institute Food and Nutrition, P.O. Box 557, 6700 AN Wageningen, The Netherlands
| | - Karen van Eunen
- Center for Liver, Digestive and Metabolic Diseases, Department of Pediatrics & Systems Biology Center for Energy Metabolism and Ageing, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Netherlands Consortium for Systems Biology, Amsterdam, The Netherlands
- Top Institute Food and Nutrition, P.O. Box 557, 6700 AN Wageningen, The Netherlands
| | - Rick Havinga
- Center for Liver, Digestive and Metabolic Diseases, Department of Pediatrics & Systems Biology Center for Energy Metabolism and Ageing, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Albert K. Groen
- Center for Liver, Digestive and Metabolic Diseases, Department of Pediatrics & Systems Biology Center for Energy Metabolism and Ageing, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Netherlands Consortium for Systems Biology, Amsterdam, The Netherlands
- Top Institute Food and Nutrition, P.O. Box 557, 6700 AN Wageningen, The Netherlands
| | - Dirk-Jan Reijngoud
- Center for Liver, Digestive and Metabolic Diseases, Department of Pediatrics & Systems Biology Center for Energy Metabolism and Ageing, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Netherlands Consortium for Systems Biology, Amsterdam, The Netherlands
- Top Institute Food and Nutrition, P.O. Box 557, 6700 AN Wageningen, The Netherlands
| | - Barbara M. Bakker
- Center for Liver, Digestive and Metabolic Diseases, Department of Pediatrics & Systems Biology Center for Energy Metabolism and Ageing, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Netherlands Consortium for Systems Biology, Amsterdam, The Netherlands
- Top Institute Food and Nutrition, P.O. Box 557, 6700 AN Wageningen, The Netherlands
- * E-mail:
| |
Collapse
|
3160
|
Etxeberria U, Arias N, Boqué N, Macarulla MT, Portillo MP, Milagro FI, Martinez JA. Shifts in microbiota species and fermentation products in a dietary model enriched in fat and sucrose. Benef Microbes 2015; 6:97-111. [PMID: 25213025 DOI: 10.3920/bm2013.0097] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The gastrointestinal tract harbours a 'superorganism' called the gut microbiota, which is known to play a crucial role in the onset and development of diverse diseases. This internal ecosystem, far from being a static environment, can be manipulated by diet and dietary components. Feeding animals with high-fat sucrose (HFS) diets entails diet-induced obesity, a model which is usually used in research to mimic the obese phenotype of Western societies. The aim of the present study was to identify gut microbiota dysbiosis and associated metabolic changes produced in male Wistar rats fed a HFS diet for 6 weeks and compare it with the basal microbial composition. For this purpose, DNA extracted from faeces at baseline and after treatment was analysed by amplification of the V4-V6 region of the 16S ribosomal DNA (rDNA) gene using 454 pyrosequencing. Short-chain fatty acids, i.e. acetate, propionate and butyrate, were also evaluated by gas chromatography-mass spectrometry. At the end of the treatment, gut microbiota composition significantly differed at phylum level (Firmicutes, Bacteroidetes and Proteobacteria) and class level (Erisypelotrichi, Deltaproteobacteria, Bacteroidia and Bacilli). Interestingly, the class Clostridia showed a significant decrease after HFS diet treatment, which correlated with visceral adipose tissue, and is likely mediated by dietary carbohydrates. Of particular interest, Clostridium cluster XIVa species were significantly reduced and changes were identified in the relative abundance of other specific bacterial species (Mitsuokella jalaludinii, Eubacterium ventriosum, Clostridium sp. FCB90-3, Prevotella nanceiensis, Clostridium fusiformis, Clostridium sp. BNL1100 and Eubacterium cylindroides) that, in some cases, showed opposite trends to their relative families. These results highlight the relevance of characterising gut microbial population differences at species level and contribute to understand the plausible link between diet and specific gut bacterial species that are able to influence the inflammatory status, intestinal barrier function and obesity development.
Collapse
Affiliation(s)
- U Etxeberria
- Department of Nutrition, Food Science and Physiology, University of Navarra, C/Irunlarrea s/n, 31008 Pamplona, Spain Centre for Nutrition Research, University of Navarra, Irunlarrea St. E-31008 Pamplona, Spain
| | - N Arias
- Nutrition and Obesity group, Department of Nutrition and Food Sciences, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria, Spain
| | - N Boqué
- Nutrition and Health Research Group. Technological Center of Nutrition and Health (CTNS), TECNIO, CEIC S. Avinguda Universitat, 1, 43204 Reus, Spain
| | - M T Macarulla
- Nutrition and Obesity group, Department of Nutrition and Food Sciences, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria, Spain CIBERobn Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - M P Portillo
- Nutrition and Obesity group, Department of Nutrition and Food Sciences, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria, Spain CIBERobn Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - F I Milagro
- Department of Nutrition, Food Science and Physiology, University of Navarra, C/Irunlarrea s/n, 31008 Pamplona, Spain Centre for Nutrition Research, University of Navarra, Irunlarrea St. E-31008 Pamplona, Spain CIBERobn Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - J A Martinez
- Department of Nutrition, Food Science and Physiology, University of Navarra, C/Irunlarrea s/n, 31008 Pamplona, Spain Centre for Nutrition Research, University of Navarra, Irunlarrea St. E-31008 Pamplona, Spain CIBERobn Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
3161
|
Abstract
PURPOSE OF REVIEW The purpose of this review was to highlight recent research developments on effects of the dietary fibre polydextrose (PDX) on appetite, satiety and energy intake and glucose metabolism. For this purpose, clinically relevant human studies were reviewed and putative mechanisms and pathways were discussed. RECENT FINDINGS A number of acute human intervention studies provide strong indications for an energy and glucose metabolism-regulating role of PDX. These effects might be mediated via a reduced gastro-intestinal transit reducing glycaemia and insulinemia after PDX ingestion and the potential of PDX as soluble dietary fibre to alter the intestinal microbial composition, which might lead to changes in signalling in both peripheral and central pathways involved in energy metabolism and glucose homeostasis. SUMMARY In acute studies, PDX seems to have an inhibiting effect on energy intake and satiety and to reduce glycaemic and insulinemic response through effect on gastro-intestinal transit time and macronutrient absorption as well as through effects of the microbial products such as short-chain fatty acids on energy and substrate metabolism. In particular, well controlled human intervention studies are required to confirm these effects in the long term. Overall, supplement PDX to the daily diet may be a promising approach for the management and treatment of obesity and associated metabolic disorders.
Collapse
Affiliation(s)
- Emanuel E Canfora
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, The Netherlands
| | | |
Collapse
|
3162
|
den Besten G, Bleeker A, Gerding A, van Eunen K, Havinga R, van Dijk TH, Oosterveer MH, Jonker JW, Groen AK, Reijngoud DJ, Bakker BM. Short-Chain Fatty Acids Protect Against High-Fat Diet-Induced Obesity via a PPARγ-Dependent Switch From Lipogenesis to Fat Oxidation. Diabetes 2015; 64:2398-408. [PMID: 25695945 DOI: 10.2337/db14-1213] [Citation(s) in RCA: 776] [Impact Index Per Article: 77.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 02/09/2015] [Indexed: 12/20/2022]
Abstract
Short-chain fatty acids (SCFAs) are the main products of dietary fiber fermentation and are believed to drive the fiber-related prevention of the metabolic syndrome. Here we show that dietary SCFAs induce a peroxisome proliferator-activated receptor-γ (PPARγ)-dependent switch from lipid synthesis to utilization. Dietary SCFA supplementation prevented and reversed high-fat diet-induced metabolic abnormalities in mice by decreasing PPARγ expression and activity. This increased the expression of mitochondrial uncoupling protein 2 and raised the AMP-to-ATP ratio, thereby stimulating oxidative metabolism in liver and adipose tissue via AMPK. The SCFA-induced reduction in body weight and stimulation of insulin sensitivity were absent in mice with adipose-specific disruption of PPARγ. Similarly, SCFA-induced reduction of hepatic steatosis was absent in mice lacking hepatic PPARγ. These results demonstrate that adipose and hepatic PPARγ are critical mediators of the beneficial effects of SCFAs on the metabolic syndrome, with clearly distinct and complementary roles. Our findings indicate that SCFAs may be used therapeutically as cheap and selective PPARγ modulators.
Collapse
Affiliation(s)
- Gijs den Besten
- Center for Liver, Digestive and Metabolic Diseases, Department of Pediatrics and Systems Biology, Center for Energy Metabolism and Ageing, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands Netherlands Consortium for Systems Biology, Amsterdam, the Netherlands
| | - Aycha Bleeker
- Center for Liver, Digestive and Metabolic Diseases, Department of Pediatrics and Systems Biology, Center for Energy Metabolism and Ageing, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands Top Institute Food and Nutrition, Wageningen, the Netherlands
| | - Albert Gerding
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Karen van Eunen
- Center for Liver, Digestive and Metabolic Diseases, Department of Pediatrics and Systems Biology, Center for Energy Metabolism and Ageing, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands Netherlands Consortium for Systems Biology, Amsterdam, the Netherlands Top Institute Food and Nutrition, Wageningen, the Netherlands
| | - Rick Havinga
- Center for Liver, Digestive and Metabolic Diseases, Department of Pediatrics and Systems Biology, Center for Energy Metabolism and Ageing, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Theo H van Dijk
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Maaike H Oosterveer
- Center for Liver, Digestive and Metabolic Diseases, Department of Pediatrics and Systems Biology, Center for Energy Metabolism and Ageing, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Johan W Jonker
- Center for Liver, Digestive and Metabolic Diseases, Department of Pediatrics and Systems Biology, Center for Energy Metabolism and Ageing, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Albert K Groen
- Center for Liver, Digestive and Metabolic Diseases, Department of Pediatrics and Systems Biology, Center for Energy Metabolism and Ageing, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands Netherlands Consortium for Systems Biology, Amsterdam, the Netherlands Top Institute Food and Nutrition, Wageningen, the Netherlands Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Dirk-Jan Reijngoud
- Center for Liver, Digestive and Metabolic Diseases, Department of Pediatrics and Systems Biology, Center for Energy Metabolism and Ageing, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands Netherlands Consortium for Systems Biology, Amsterdam, the Netherlands Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Barbara M Bakker
- Center for Liver, Digestive and Metabolic Diseases, Department of Pediatrics and Systems Biology, Center for Energy Metabolism and Ageing, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands Netherlands Consortium for Systems Biology, Amsterdam, the Netherlands
| |
Collapse
|
3163
|
Correlation of Lactobacillus rhamnosus Genotypes and Carbohydrate Utilization Signatures Determined by Phenotype Profiling. Appl Environ Microbiol 2015; 81:5458-70. [PMID: 26048937 DOI: 10.1128/aem.00851-15] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2015] [Accepted: 05/28/2015] [Indexed: 12/17/2022] Open
Abstract
Lactobacillus rhamnosus is a bacterial species commonly colonizing the gastrointestinal (GI) tract of humans and also frequently used in food products. While some strains have been studied extensively, physiological variability among isolates of the species found in healthy humans or their diet is largely unexplored. The aim of this study was to characterize the diversity of carbohydrate utilization capabilities of human isolates and food-derived strains of L. rhamnosus in relation to their niche of isolation and genotype. We investigated the genotypic and phenotypic diversity of 25 out of 65 L. rhamnosus strains from various niches, mainly human feces and fermented dairy products. Genetic fingerprinting of the strains by amplified fragment length polymorphism (AFLP) identified 11 distinct subgroups at 70% similarity and suggested niche enrichment within particular genetic clades. High-resolution carbohydrate utilization profiling (OmniLog) identified 14 carbon sources that could be used by all of the strains tested for growth, while the utilization of 58 carbon sources differed significantly between strains, enabling the stratification of L. rhamnosus strains into three metabolic clusters that partially correlate with the genotypic clades but appear uncorrelated with the strain's origin of isolation. Draft genome sequences of 8 strains were generated and employed in a gene-trait matching (GTM) analysis together with the publicly available genomes of L. rhamnosus GG (ATCC 53103) and HN001 for several carbohydrates that were distinct for the different metabolic clusters: l-rhamnose, cellobiose, l-sorbose, and α-methyl-d-glucoside. From the analysis, candidate genes were identified that correlate with l-sorbose and α-methyl-d-glucoside utilization, and the proposed function of these genes could be confirmed by heterologous expression in a strain lacking the genes. This study expands our insight into the phenotypic and genotypic diversity of the species L. rhamnosus and explores the relationships between specific carbohydrate utilization capacities and genotype and/or niche adaptation of this species.
Collapse
|
3164
|
Aryana K, Greenway F, Dhurandhar N, Tulley R, Finley J, Keenan M, Martin R, Pelkman C, Olson D, Zheng J. A resistant-starch enriched yogurt: fermentability, sensory characteristics, and a pilot study in children. F1000Res 2015; 4:139. [PMID: 26925221 PMCID: PMC4712773 DOI: 10.12688/f1000research.6451.1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/19/2015] [Indexed: 12/11/2022] Open
Abstract
The rising prevalence of obesity and the vulnerability of the pediatric age group have highlighted the critical need for a careful consideration of effective, safe, remedial and preventive dietary interventions. Amylose starch (RS2) from high-amylose maize (HAM) ferments in the gut and affects body weight. One hundred and ten children, of 7-8 (n=91) or 13-14 (n=19) years of age scored the sensory qualities of a yogurt supplemented with either HAM-RS2 or an amylopectin starch. The amylopectin starch yogurt was preferred to the HAM-RS2-enriched yogurt by 7-8 year old panelists (
P<0.0001). Appearance, taste, and sandiness scores given by 13- to 14-year-old panelists were more favorable for the amylopectin starch yogurt than for HAM-RS2-enriched yogurt (
P<0.05). HAM-RS2 supplementation resulted in acceptable (≥6 on a 1-9 scale) sensory and hedonic ratings of the yogurt in 74% of subjects. Four children consumed a HAM-RS2-enriched yogurt for four weeks to test its fermentability in a clinical trial. Three adolescents, but not the single pre-pubertal child, had reduced stool pH (
P=0.1) and increased stool short-chain fatty acids (SCFAs) (
P<0.05) including increased fecal acetate (
P=0.02), and butyrate (
P=0.089) from resistant starch (RS) fermentation and isobutyrate (
P=0.01) from protein fermentation post-treatment suggesting a favorable change to the gut microbiota. HAM-RS2 was not modified by pasteurization of the yogurt, and may be a palatable way to increase fiber intake and stimulate colonic fermentation in adolescents. Future studies are planned to determine the concentration of HAM-RS2 that offers the optimal safe and effective strategy to prevent excessive fat gain in children.
Collapse
Affiliation(s)
- Kayanush Aryana
- School of Animal Sciences, College of Agriculture, Louisiana State University, Baton Rouge, LA, 70808, USA; Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, 70808, USA
| | - Frank Greenway
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, 70808, USA
| | - Nikhil Dhurandhar
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, 79409, USA
| | - Richard Tulley
- School of Nutrition and Food Science, College of Agriculture, Louisiana State University, Baton Rouge, LA, 70803, USA
| | - John Finley
- School of Nutrition and Food Science, College of Agriculture, Louisiana State University, Baton Rouge, LA, 70803, USA
| | - Michael Keenan
- School of Nutrition and Food Science, College of Agriculture, Louisiana State University, Baton Rouge, LA, 70803, USA
| | - Roy Martin
- University of California, University of California, Davis, CA, 95616, USA
| | | | - Douglas Olson
- School of Animal Sciences, College of Agriculture, Louisiana State University, Baton Rouge, LA, 70808, USA
| | - Jolene Zheng
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, 70808, USA
| |
Collapse
|
3165
|
Han NE, Lee EJ, Park KS, Jeon IS, Lee HK, Song KD, Choi JK. Analysis of porcine macrophage immune response to antigenic molecules and short chain fatty acids. J Biomed Res 2015. [DOI: 10.12729/jbr.2015.16.2.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
3166
|
Verbeke KA, Boobis AR, Chiodini A, Edwards CA, Franck A, Kleerebezem M, Nauta A, Raes J, van Tol EAF, Tuohy KM, on behalf of the ILSI Europe Prebiotics Task Force Expert Group ‘Microbial metabolism and fermentation’. Towards microbial fermentation metabolites as markers for health benefits of prebiotics. Nutr Res Rev 2015; 28:42-66. [PMID: 26156216 PMCID: PMC4501371 DOI: 10.1017/s0954422415000037] [Citation(s) in RCA: 225] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Available evidence on the bioactive, nutritional and putative detrimental properties of gut microbial metabolites has been evaluated to support a more integrated view of how prebiotics might affect host health throughout life. The present literature inventory targeted evidence for the physiological and nutritional effects of metabolites, for example, SCFA, the potential toxicity of other metabolites and attempted to determine normal concentration ranges. Furthermore, the biological relevance of more holistic approaches like faecal water toxicity assays and metabolomics and the limitations of faecal measurements were addressed. Existing literature indicates that protein fermentation metabolites (phenol, p-cresol, indole, ammonia), typically considered as potentially harmful, occur at concentration ranges in the colon such that no toxic effects are expected either locally or following systemic absorption. The endproducts of saccharolytic fermentation, SCFA, may have effects on colonic health, host physiology, immunity, lipid and protein metabolism and appetite control. However, measuring SCFA concentrations in faeces is insufficient to assess the dynamic processes of their nutrikinetics. Existing literature on the usefulness of faecal water toxicity measures as indicators of cancer risk seems limited. In conclusion, at present there is insufficient evidence to use changes in faecal bacterial metabolite concentrations as markers of prebiotic effectiveness. Integration of results from metabolomics and metagenomics holds promise for understanding the health implications of prebiotic microbiome modulation but adequate tools for data integration and interpretation are currently lacking. Similarly, studies measuring metabolite fluxes in different body compartments to provide a more accurate picture of their nutrikinetics are needed.
Collapse
Affiliation(s)
- Kristin A. Verbeke
- Translational Research in Gastrointestinal Disorders (TARGID), KU Leuven and Leuven Food Science and Nutrition Research Center (LFoRCe), Leuven, Belgium
| | - Alan R. Boobis
- Department of Medicine, Imperial College London, London, UK
| | - Alessandro Chiodini
- Formerly ILSI Europe, Box 6, Avenue Emmanuel Mounier 83, BE-1200, Brussels, Belgium; now European Commission, Research Executive Agency (REA) Unit B2, Brussels, Belgium
| | - Christine A. Edwards
- Human Nutrition School of Medicine, College of MVLS, University of Glasgow, Glasgow, Scotland
| | | | - Michiel Kleerebezem
- Host Microbe Interactomics, Wageningen University, Wageningen, The Netherlands
| | - Arjen Nauta
- FrieslandCampina, Amersfoort, The Netherlands
| | - Jeroen Raes
- Microbiology and Immunology, Rega Institute, KU Leuven, Leuven; VIB, Leuven; DBIT, Vrije Universiteit Brussel, Brussels, Belgium
| | | | - Kieran M. Tuohy
- Nutrition and Nutrigenomics, Research and Innovation Centre-Fondazione Edmund Mach, Trento, Italy
| | | |
Collapse
|
3167
|
Cooper DN, Martin RJ, Keim NL. Does Whole Grain Consumption Alter Gut Microbiota and Satiety? Healthcare (Basel) 2015; 3:364-92. [PMID: 27417768 PMCID: PMC4939539 DOI: 10.3390/healthcare3020364] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 05/19/2015] [Accepted: 05/22/2015] [Indexed: 12/25/2022] Open
Abstract
This review summarizes recent studies examining whole grain consumption and its effect on gut microbiota and satiety in healthy humans. Studies comparing whole grains to their refined grain counterparts were considered, as were studies comparing different grain types. Possible mechanisms linking microbial metabolism and satiety are described. Clinical trials show that whole grain wheat, maize, and barley alter the human gut microbiota, but these findings are based on a few studies that do not include satiety components, so no functional claims between microbiota and satiety can be made. Ten satiety trials were evaluated and provide evidence that whole oats, barley, and rye can increase satiety, whereas the evidence for whole wheat and maize is not compelling. There are many gaps in the literature; no one clinical trial has examined the effects of whole grains on satiety and gut microbiota together. Once understanding the impact of whole grains on satiety and microbiota is more developed, then particular grains might be used for better appetite control. With this information at hand, healthcare professionals could make individual dietary recommendations that promote satiety and contribute to weight control.
Collapse
Affiliation(s)
- Danielle N Cooper
- Department of Nutrition, University of California at Davis, 1 Shields Ave, Davis, CA 95616, USA.
| | - Roy J Martin
- Department of Nutrition, University of California at Davis, 1 Shields Ave, Davis, CA 95616, USA.
- USDA-ARS, Western Human Nutrition Research Center, 430 West Health Sciences Drive, Davis, CA 95616, USA.
| | - Nancy L Keim
- Department of Nutrition, University of California at Davis, 1 Shields Ave, Davis, CA 95616, USA.
- USDA-ARS, Western Human Nutrition Research Center, 430 West Health Sciences Drive, Davis, CA 95616, USA.
| |
Collapse
|
3168
|
Metagenome Sequencing of the Hadza Hunter-Gatherer Gut Microbiota. Curr Biol 2015; 25:1682-93. [PMID: 25981789 DOI: 10.1016/j.cub.2015.04.055] [Citation(s) in RCA: 254] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 04/09/2015] [Accepted: 04/29/2015] [Indexed: 12/21/2022]
Abstract
Through human microbiome sequencing, we can better understand how host evolutionary and ontogenetic history is reflected in the microbial function. However, there has been no information on the gut metagenome configuration in hunter-gatherer populations, posing a gap in our knowledge of gut microbiota (GM)-host mutualism arising from a lifestyle that describes over 90% of human evolutionary history. Here, we present the first metagenomic analysis of GM from Hadza hunter-gatherers of Tanzania, showing a unique enrichment in metabolic pathways that aligns with the dietary and environmental factors characteristic of their foraging lifestyle. We found that the Hadza GM is adapted for broad-spectrum carbohydrate metabolism, reflecting the complex polysaccharides in their diet. Furthermore, the Hadza GM is equipped for branched-chain amino acid degradation and aromatic amino acid biosynthesis. Resistome functionality demonstrates the existence of antibiotic resistance genes in a population with little antibiotic exposure, indicating the ubiquitous presence of environmentally derived resistances. Our results demonstrate how the functional specificity of the GM correlates with certain environment and lifestyle factors and how complexity from the exogenous environment can be balanced by endogenous homeostasis. The Hadza gut metagenome structure allows us to appreciate the co-adaptive functional role of the GM in complementing the human physiology, providing a better understanding of the versatility of human life and subsistence.
Collapse
|
3169
|
Frye RE, Rose S, Slattery J, MacFabe DF. Gastrointestinal dysfunction in autism spectrum disorder: the role of the mitochondria and the enteric microbiome. MICROBIAL ECOLOGY IN HEALTH AND DISEASE 2015; 26:27458. [PMID: 25956238 PMCID: PMC4425813 DOI: 10.3402/mehd.v26.27458] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Revised: 04/09/2015] [Accepted: 04/10/2015] [Indexed: 12/26/2022]
Abstract
Autism spectrum disorder (ASD) affects a significant number of individuals worldwide with the prevalence continuing to grow. It is becoming clear that a large subgroup of individuals with ASD demonstrate abnormalities in mitochondrial function as well as gastrointestinal (GI) symptoms. Interestingly, GI disturbances are common in individuals with mitochondrial disorders and have been reported to be highly prevalent in individuals with co-occurring ASD and mitochondrial disease. The majority of individuals with ASD and mitochondrial disorders do not manifest a primary genetic mutation, raising the possibility that their mitochondrial disorder is acquired or, at least, results from a combination of genetic susceptibility interacting with a wide range of environmental triggers. Mitochondria are very sensitive to both endogenous and exogenous environmental stressors such as toxicants, iatrogenic medications, immune activation, and metabolic disturbances. Many of these same environmental stressors have been associated with ASD, suggesting that the mitochondria could be the biological link between environmental stressors and neurometabolic abnormalities associated with ASD. This paper reviews the possible links between GI abnormalities, mitochondria, and ASD. First, we review the link between GI symptoms and abnormalities in mitochondrial function. Second, we review the evidence supporting the notion that environmental stressors linked to ASD can also adversely affect both mitochondria and GI function. Third, we review the evidence that enteric bacteria that are overrepresented in children with ASD, particularly Clostridia spp., produce short-chain fatty acid metabolites that are potentially toxic to the mitochondria. We provide an example of this gut–brain connection by highlighting the propionic acid rodent model of ASD and the clinical evidence that supports this animal model. Lastly, we discuss the potential therapeutic approaches that could be helpful for GI symptoms in ASD and mitochondrial disorders. To this end, this review aims to help better understand the underlying pathophysiology associated with ASD that may be related to concurrent mitochondrial and GI dysfunction.
Collapse
Affiliation(s)
- Richard E Frye
- Autism Research Program, Arkansas Children's Hospital Research Institute, Little Rock, AR, USA.,Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA;
| | - Shannon Rose
- Autism Research Program, Arkansas Children's Hospital Research Institute, Little Rock, AR, USA.,Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - John Slattery
- Autism Research Program, Arkansas Children's Hospital Research Institute, Little Rock, AR, USA.,Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Derrick F MacFabe
- Kilee Patchell-Evans Autism Research Group, Division of Developmental Disabilities, Departments of Psychology and Psychiatry, University of Western Ontario, London, ON, Canada
| |
Collapse
|
3170
|
Di Stasi LC, Costa CA, Witaicenis A. Products for the treatment of inflammatory bowel disease: a patent review (2013 - 2014). Expert Opin Ther Pat 2015; 25:629-42. [PMID: 25944127 DOI: 10.1517/13543776.2015.1041921] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Inflammatory bowel disease (IBD) consists of Crohn's disease, ulcerative colitis and an unspecific IBD. The unclear etiology of IBD is a limiting factor that complicates the development of new pharmacological treatments and explains the high frequency of refractory patients to current drugs, including both conventional and biological therapies. In view of this, recent progress on the development of novel patented products to treat IBD was reviewed. AREAS COVERED Evaluation of the patent literature during the period 2013 - 2014 focused on chemical compounds, functional foods and biological therapy useful for the treatment of IBD. EXPERT OPINION Majority of the patents are not conclusive because they were based on data from unspecific methods not related to intestinal inflammation and, when related to IBD models, few biochemical and molecular evaluations that could be corroborating their use in human IBD were presented. On the other hand, methods and strategies using new formulations of conventional drugs, guanylyl cyclase C peptide agonists, compounds that influence anti-adhesion molecules, mAbs anti-type I interferons and anti-integrin, oligonucleotide antisense Smad7, growth factor neuregulin 4 and functional foods, particularly fermented wheat germ with Saccharomyces cerevisiae, are promising products for use in the very near future.
Collapse
Affiliation(s)
- Luiz C Di Stasi
- São Paulo State University (UNESP), Institute of Biosciences, Department of Pharmacology, Laboratory of Phytomedicines, Pharmacology and Biotechnology (PhytoPharmaTech) , 18.618-000 - Botucatu, São Paulo , Brazil +55 14 3880 0216 ;
| | | | | |
Collapse
|
3171
|
Houlden A, Hayes KS, Bancroft AJ, Worthington JJ, Wang P, Grencis RK, Roberts IS. Chronic Trichuris muris Infection in C57BL/6 Mice Causes Significant Changes in Host Microbiota and Metabolome: Effects Reversed by Pathogen Clearance. PLoS One 2015; 10:e0125945. [PMID: 25938477 PMCID: PMC4418675 DOI: 10.1371/journal.pone.0125945] [Citation(s) in RCA: 155] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 03/25/2015] [Indexed: 12/26/2022] Open
Abstract
Trichuris species are a globally important and prevalent group of intestinal helminth parasites, in which Trichuris muris (mouse whipworm) is an ideal model for this disease. This paper describes the first ever highly controlled and comprehensive investigation into the effects of T. muris infection on the faecal microbiota of mice and the effects on the microbiota following successful clearance of the infection. Communities were profiled using DGGE, 454 pyrosequencing, and metabolomics. Changes in microbial composition occurred between 14 and 28 days post infection, resulting in significant changes in α and β- diversity. This impact was dominated by a reduction in the diversity and abundance of Bacteroidetes, specifically Prevotella and Parabacteroides. Metabolomic analysis of stool samples of infected mice at day 41 showed significant differences to uninfected controls with a significant increase in the levels of a number of essential amino acids and a reduction in breakdown of dietary plant derived carbohydrates. The significant reduction in weight gain by infected mice probably reflects these metabolic changes and the incomplete digestion of dietary polysaccharides. Following clearance of infection the intestinal microbiota underwent additional changes gradually transitioning by day 91 towards a microbiota of an uninfected animal. These data indicate that the changes in microbiota as a consequence of infection were transitory requiring the presence of the pathogen for maintenance. Interestingly this was not observed for all of the key immune cell populations associated with chronic T. muris infection. This reflects the highly regulated chronic response and potential lasting immunological consequences of dysbiosis in the microbiota. Thus infection of T. muris causes a significant and substantial impact on intestinal microbiota and digestive function of mice with affects in long term immune regulation.
Collapse
Affiliation(s)
- Ashley Houlden
- Faculty of Life Sciences, University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Kelly S. Hayes
- Faculty of Life Sciences, University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Allison J. Bancroft
- Faculty of Life Sciences, University of Manchester, Manchester, M13 9PT, United Kingdom
| | - John J. Worthington
- Faculty of Life Sciences, University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Ping Wang
- Faculty of Life Sciences, University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Richard K. Grencis
- Faculty of Life Sciences, University of Manchester, Manchester, M13 9PT, United Kingdom
- * E-mail: (RKG); (ISR)
| | - Ian S. Roberts
- Faculty of Life Sciences, University of Manchester, Manchester, M13 9PT, United Kingdom
- * E-mail: (RKG); (ISR)
| |
Collapse
|
3172
|
Potgieter M, Bester J, Kell DB, Pretorius E. The dormant blood microbiome in chronic, inflammatory diseases. FEMS Microbiol Rev 2015; 39:567-91. [PMID: 25940667 PMCID: PMC4487407 DOI: 10.1093/femsre/fuv013] [Citation(s) in RCA: 288] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/02/2015] [Indexed: 02/07/2023] Open
Abstract
Blood in healthy organisms is seen as a ‘sterile’ environment: it lacks proliferating microbes. Dormant or not-immediately-culturable forms are not absent, however, as intracellular dormancy is well established. We highlight here that a great many pathogens can survive in blood and inside erythrocytes. ‘Non-culturability’, reflected by discrepancies between plate counts and total counts, is commonplace in environmental microbiology. It is overcome by improved culturing methods, and we asked how common this would be in blood. A number of recent, sequence-based and ultramicroscopic studies have uncovered an authentic blood microbiome in a number of non-communicable diseases. The chief origin of these microbes is the gut microbiome (especially when it shifts composition to a pathogenic state, known as ‘dysbiosis’). Another source is microbes translocated from the oral cavity. ‘Dysbiosis’ is also used to describe translocation of cells into blood or other tissues. To avoid ambiguity, we here use the term ‘atopobiosis’ for microbes that appear in places other than their normal location. Atopobiosis may contribute to the dynamics of a variety of inflammatory diseases. Overall, it seems that many more chronic, non-communicable, inflammatory diseases may have a microbial component than are presently considered, and may be treatable using bactericidal antibiotics or vaccines. Atopobiosis of microbes (the term describing microbes that appear in places other than where they should be), as well as the products of their metabolism, seems to correlate with, and may contribute to, the dynamics of a variety of inflammatory diseases.
Collapse
Affiliation(s)
- Marnie Potgieter
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Arcadia 0007, South Africa
| | - Janette Bester
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Arcadia 0007, South Africa
| | - Douglas B Kell
- School of Chemistry and The Manchester Institute of Biotechnology, The University of Manchester, 131, Princess St, Manchester M1 7DN, Lancs, UK
| | - Etheresia Pretorius
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Arcadia 0007, South Africa
| |
Collapse
|
3173
|
Aguirre M, Venema K. Does the Gut Microbiota Contribute to Obesity? Going beyond the Gut Feeling. Microorganisms 2015; 3:213-35. [PMID: 27682087 PMCID: PMC5023237 DOI: 10.3390/microorganisms3020213] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Revised: 04/05/2015] [Accepted: 04/17/2015] [Indexed: 02/06/2023] Open
Abstract
Increasing evidence suggests that gut microbiota is an environmental factor that plays a crucial role in obesity. However, the aetiology of obesity is rather complex and depends on different factors. Furthermore, there is a lack of consensus about the exact role that this microbial community plays in the host. The aim of this review is to present evidence about what has been characterized, compositionally and functionally, as obese gut microbiota. In addition, the different reasons explaining the so-far unclear role are discussed considering evidence from in vitro, animal and human studies.
Collapse
Affiliation(s)
- Marisol Aguirre
- Top Institute of Food and Nutrition, P.O. Box 557, 6700 AA Wageningen, The Netherlands.
- School of Nutrition and Translational Research in Metabolism (NUTRIM), Faculty of Health, Medicine and Life Sciences, Department of Human Biology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands.
- The Netherlands Organization for Applied Scientific Research (TNO), P.O. Box 360, 3700 AJ Zeist, The Netherlands.
| | - Koen Venema
- Top Institute of Food and Nutrition, P.O. Box 557, 6700 AA Wageningen, The Netherlands.
- Beneficial Microbes Consultancy, Johan Karschstraat 3, 6709 TN Wageningen, The Netherlands.
| |
Collapse
|
3174
|
Dietary gut microbial metabolites, short-chain fatty acids, and host metabolic regulation. Nutrients 2015; 7:2839-49. [PMID: 25875123 PMCID: PMC4425176 DOI: 10.3390/nu7042839] [Citation(s) in RCA: 592] [Impact Index Per Article: 59.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 03/29/2015] [Accepted: 04/02/2015] [Indexed: 02/06/2023] Open
Abstract
During feeding, the gut microbiota contributes to the host energy acquisition and metabolic regulation thereby influencing the development of metabolic disorders such as obesity and diabetes. Short-chain fatty acids (SCFAs) such as acetate, butyrate, and propionate, which are produced by gut microbial fermentation of dietary fiber, are recognized as essential host energy sources and act as signal transduction molecules via G-protein coupled receptors (FFAR2, FFAR3, OLFR78, GPR109A) and as epigenetic regulators of gene expression by the inhibition of histone deacetylase (HDAC). Recent evidence suggests that dietary fiber and the gut microbial-derived SCFAs exert multiple beneficial effects on the host energy metabolism not only by improving the intestinal environment, but also by directly affecting various host peripheral tissues. In this review, we summarize the roles of gut microbial SCFAs in the host energy regulation and present an overview of the current understanding of its physiological functions.
Collapse
|
3175
|
Graham C, Mullen A, Whelan K. Obesity and the gastrointestinal microbiota: a review of associations and mechanisms. Nutr Rev 2015; 73:376-85. [PMID: 26011912 DOI: 10.1093/nutrit/nuv004] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The two-way obesity model that considers only the interplay between humans and their environment has been revised to include the gastrointestinal microbiota. Notable perturbations in the bacterial communities in obese individuals have been uncovered. Research is helping to distinguish between the obesogenic mechanisms attributable to diet and those that may be associated with the microbiota. Examples include studies in which transplant of the microbiota from murine models of weight loss (gastric bypass) into germ-free mice resulted in significant weight loss. Several mechanisms have been identified that suggest the microbiota may play a role in obesity development and propagation. There is some evidence from animal and human studies that the microbiota in the obese harvests energy more effectively and may manipulate host gene function leading to increased adiposity, aggravation of inflammatory mechanisms, metabolic endotoxemia, and metabolic dysfunction. Research findings highlight the potential of the microbiota to influence body weight and they allude to its potential therapeutic use in tackling the costly global epidemic of obesity.
Collapse
Affiliation(s)
- Catherine Graham
- C. Graham, A. Mullen, and K. Whelan are with the Diabetes and Nutritional Sciences Division, Faculty of Life Sciences & Medicine, King's College London, London, SE1 9NN, UK
| | - Anne Mullen
- C. Graham, A. Mullen, and K. Whelan are with the Diabetes and Nutritional Sciences Division, Faculty of Life Sciences & Medicine, King's College London, London, SE1 9NN, UK
| | - Kevin Whelan
- C. Graham, A. Mullen, and K. Whelan are with the Diabetes and Nutritional Sciences Division, Faculty of Life Sciences & Medicine, King's College London, London, SE1 9NN, UK.
| |
Collapse
|
3176
|
Heimann E, Nyman M, Degerman E. Propionic acid and butyric acid inhibit lipolysis and de novo lipogenesis and increase insulin-stimulated glucose uptake in primary rat adipocytes. Adipocyte 2015; 4:81-8. [PMID: 26167409 PMCID: PMC4496978 DOI: 10.4161/21623945.2014.960694] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Revised: 07/10/2014] [Accepted: 08/28/2014] [Indexed: 01/21/2023] Open
Abstract
Fermentation of dietary fibers by colonic microbiota generates short-chain fatty acids (SCFAs), e.g., propionic acid and butyric acid, which have been described to have “anti-obesity properties” by ameliorating fasting glycaemia, body weight and insulin tolerance in animal models. In the present study, we therefore investigate if propionic acid and butyric acid have effects on lipolysis, de novo lipogenesis and glucose uptake in primary rat adipocytes. We show that both propionic acid and butyric acid inhibit isoproterenol- and adenosine deaminase-stimulated lipolysis as well as isoproterenol-stimulated lipolysis in the presence of a phosphodiesterase (PDE3) inhibitor. In addition, we show that propionic acid and butyric acid inhibit basal and insulin-stimulated de novo lipogenesis, which is associated with increased phosphorylation and thus inhibition of acetyl CoA carboxylase, a rate-limiting enzyme in fatty acid synthesis. Furthermore, we show that propionic acid and butyric acid increase insulin-stimulated glucose uptake. To conclude, our study shows that SCFAs have effects on fat storage and mobilization as well as glucose uptake in rat primary adipocytes. Thus, the SCFAs might contribute to healthier adipocytes and subsequently also to improved energy metabolism with for example less circulating free fatty acids, which is beneficial in the context of obesity and type 2 diabetes.
Collapse
Key Words
- ACC, acetyl-CoA carboxylase
- ADA, adenosine deaminase
- AMPK, AMP-activated protein kinase
- BA, butyric acid
- BSA, bovine serum albumin
- FFAR, free fatty acid receptor
- GLUT, glucose transporter
- GPCR, G-protein-coupled receptor
- HSL, hormone-sensitive lipase
- ISO, isoproterenol
- KRBH, Krebs-Ringer bicarbonate-HEPES
- KRH, Krebs Ringer-HEPES
- PA, propionic acid
- PDE, cyclic nucleotide phosphodiesterase
- SCFAs, short-chain fatty acids
- T2D, type 2 diabetes
- adipocyte
- metabolism
- obesity
- short-chain fatty acid
- type 2 diabetes
Collapse
|
3177
|
Heinzmann SS, Schmitt-Kopplin P. Deep Metabotyping of the Murine Gastrointestinal Tract for the Visualization of Digestion and Microbial Metabolism. J Proteome Res 2015; 14:2267-77. [DOI: 10.1021/acs.jproteome.5b00034] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Silke S. Heinzmann
- Research
Unit Analytical BioGeoChemistry, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Philippe Schmitt-Kopplin
- Research
Unit Analytical BioGeoChemistry, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
- Chair
of Analytical Food Chemistry, Technische Universität München, 85354 Freising, Germany
| |
Collapse
|
3178
|
Koleva PT, Bridgman SL, Kozyrskyj AL. The infant gut microbiome: evidence for obesity risk and dietary intervention. Nutrients 2015; 7:2237-60. [PMID: 25835047 PMCID: PMC4425142 DOI: 10.3390/nu7042237] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 03/04/2015] [Accepted: 03/18/2015] [Indexed: 12/26/2022] Open
Abstract
Increasing globally, particularly in children, obesity is a serious public health issue and risk factor for overweight and metabolic disease in later life. Both in experimental animal and human studies, advances in gene sequencing technologies have yielded intriguing possibilities for the role of the gut microbiome in later development of overweight status. Before translating study findings into practice, we must first reconcile inconsistencies between animal experimentation, and human adult and infant studies. Recent evidence for associations with gut microbiota and infant weight gain or child weight status, implicate Bacteroides and Lactobacillus species. Dietary manipulation with human milk and pre/probiotic formulations holds promise for preventing obesity.
Collapse
Affiliation(s)
- Petya T Koleva
- Department of Pediatrics, University of Alberta, Edmonton, AB, T6G 1C9, Canada.
| | - Sarah L Bridgman
- Department of Pediatrics, University of Alberta, Edmonton, AB, T6G 1C9, Canada.
| | - Anita L Kozyrskyj
- Department of Pediatrics, University of Alberta, Edmonton, AB, T6G 1C9, Canada.
- School of Public Health, University of Alberta, Edmonton, AB, T6G 1C9, Canada.
- Community Health Sciences, University of Manitoba, Winnipeg, MB, R3E 0W3, Canada.
| |
Collapse
|
3179
|
Fröhlich EE, Mayerhofer R, Holzer P. Reevaluating the hype: four bacterial metabolites under scrutiny. Eur J Microbiol Immunol (Bp) 2015; 5:1-13. [PMID: 25883790 DOI: 10.1556/eujmi-d-14-00030] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 11/06/2014] [Indexed: 12/20/2022] Open
Abstract
With microbiome research being a fiercely contested playground in science, new data are being published at tremendous pace. The review at hand serves to critically revise four microbial metabolites widely applied in research: butyric acid, flagellin, lipoteichoic acid, and propionic acid. All four metabolites are physiologically present in healthy humans. Nevertheless, all four are likewise involved in pathologies ranging from cancer to mental retardation. Their inflammatory potential is equally friend and foe. The authors systematically analyze positive and negative attributes of the aforementioned substances, indicating chances and dangers with the use of pre- and probiotic therapeutics. Furthermore, the widespread actions of microbial metabolites on distinct organs and diseases are reconciled. Moreover, the review serves as critical discourse on scientific methods commonly employed in microbiome research and comparability as well as reproducibility issues arising thereof.
Collapse
Affiliation(s)
- E E Fröhlich
- Research Unit of Translational Neurogastroenterology, Institute of Experimental and Clinical Pharmacology, Medical University of Graz Universitätsplatz 4, 8010 Graz Austria
| | - R Mayerhofer
- Research Unit of Translational Neurogastroenterology, Institute of Experimental and Clinical Pharmacology, Medical University of Graz Universitätsplatz 4, 8010 Graz Austria
| | - P Holzer
- Research Unit of Translational Neurogastroenterology, Institute of Experimental and Clinical Pharmacology, Medical University of Graz Universitätsplatz 4, 8010 Graz Austria
| |
Collapse
|
3180
|
Involvement of the gut chemosensory system in the regulation of colonic anion secretion. BIOMED RESEARCH INTERNATIONAL 2015; 2015:403919. [PMID: 25866781 PMCID: PMC4383346 DOI: 10.1155/2015/403919] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Accepted: 09/09/2014] [Indexed: 12/26/2022]
Abstract
The primary function of the gastrointestinal (GI) tract is the extraction of nutrients from the diet. Therefore, the GI tract must possess an efficient surveillance system that continuously monitors the luminal content for beneficial or harmful compounds. Recent studies have shown that specialized cells in the intestinal lining can sense changes in this content. These changes directly influence fundamental GI processes such as secretion, motility, and local blood flow via hormonal and/or neuronal pathways. Until recently, most studies examining the control of ion transport in the colon have focused on neural and hormonal regulation. However, study of the regulation of gut function by the gut chemosensory system has become increasingly important, as failure of this system causes dysfunctions in host homeostasis, as well as functional GI disorders. Furthermore, regulation of ion transport in the colon is critical for host defense and for electrolytes balance. This review discusses the role of the gut chemosensory system in epithelial transport, with a particular emphasis on the colon.
Collapse
|
3181
|
Phylogenetic and functional alterations in bacterial community compositions in broiler ceca as a result of mannan oligosaccharide supplementation. Appl Environ Microbiol 2015; 81:3460-70. [PMID: 25769823 DOI: 10.1128/aem.04194-14] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Accepted: 03/05/2015] [Indexed: 12/20/2022] Open
Abstract
This study focused on identifying reproducible effects of dietary supplementation with a mannan oligosaccharide (MOS) on the broiler cecal bacterial community structure and function in a commercial production setting. Two separate trials, each with a control and a supplemented group, were carried out in the same commercial location and run concurrently. Approximately 10,000 birds from the same commercial hatchery were mirror imaged into each of four commercial broiler sheds and fed either a control or supplemented diet. Cecal contents were obtained on days 7, 21, and 35 posthatch from 12 randomly caught broilers from each group. Bacterial pyrosequencing was performed on all samples, with approximately 250,000 sequences obtained per treatment per time point. The predominant phyla identified at all three time points in both trials were Firmicutes, Bacteroidetes, Proteobacteria, Actinobacteria, and Tenericutes, representing >99% of all sequences. MOS supplementation altered the bacterial community composition from 7 days supplementation through 35 days supplementation. Bacteroidetes appeared to be replacing Firmicutes as a result of supplementation, with the most noticeable effects after 35 days. The effects of supplementation were reproducible across both trials. PICRUSt was used to identify differences between the functional potentials of the bacterial communities as a result of MOS supplementation. Using level 3 KEGG ortholog function predictions, differences between control and supplemented groups were observed, with very strong segregation noted on day 35 posthatch in both trials. This indicated that alterations of bacterial communities as a result of MOS are likely to alter the functional capability of the cecum.
Collapse
|
3182
|
Lu Z, Gui H, Yao L, Yan L, Martens H, Aschenbach JR, Shen Z. Short-chain fatty acids and acidic pH upregulate UT-B, GPR41, and GPR4 in rumen epithelial cells of goats. Am J Physiol Regul Integr Comp Physiol 2015; 308:R283-93. [PMID: 25519731 DOI: 10.1152/ajpregu.00323.2014] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Currently, the mechanism(s) responsible for the regulation of urea transporter B (UT-B) expression levels in the epithelium of the rumen remain unclear. We hypothesized that rumen fermentation products affect ruminal UT-B expression. Therefore, the effects of short-chain fatty acids (SCFA), pH, ammonia, and urea on mRNA and protein levels of UT-B were assayed in primary rumen epithelial cell cultures and in rumen epithelium obtained from intact goats. In vitro, SCFA and acidic pH were found to synergetically stimulate both mRNA and protein expression of UT-B, whereas NH4Cl decreased mRNA and protein levels of UT-B at pH 6.8. Treatment with urea increased both levels at pH 7.4. When goats received a diet rich in nitrogen (N) and nonfiber carbohydrates (NFC), their rumen epithelium had higher levels of UT-B, and the rumen contained higher concentrations of SCFA and NH3-N with a lower pH. An increase in plasma urea-N concentration was also observed compared with the plasma of the goats that received a diet low in N and NFC. In a second feeding trial, goats that received a NFC-rich, but isonitrogenous, diet had higher mRNA and protein levels of UT-B, and higher levels of G protein-coupled receptor (GPR) 41 and GPR4, in their rumen epithelium. The ruminal concentrations of SCFA and NH3-N also increased, while a lower pH was detected. In contrast, the serum urea-N concentrations remained unchanged. These data indicate that ruminal SCFA and pH are key factors, via GPR4 and GPR41, in the dietary regulation of UT-B expression, and they have priority over changes in plasma urea.
Collapse
Affiliation(s)
- Zhongyan Lu
- Institute of Veterinary Physiology, Free University of Berlin, Berlin, Germany; and
| | - Hongbing Gui
- Laboratory of Animal Physiology and Biochemistry, Nanjing Agricultural University, Nanjing, China
| | - Lei Yao
- Laboratory of Animal Physiology and Biochemistry, Nanjing Agricultural University, Nanjing, China
| | - Lei Yan
- Laboratory of Animal Physiology and Biochemistry, Nanjing Agricultural University, Nanjing, China
| | - Holger Martens
- Institute of Veterinary Physiology, Free University of Berlin, Berlin, Germany; and
| | - Jörg R Aschenbach
- Institute of Veterinary Physiology, Free University of Berlin, Berlin, Germany; and
| | - Zanming Shen
- Laboratory of Animal Physiology and Biochemistry, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
3183
|
Microbiota and Gut Stem Cells Cross-Talks: A New View of Epithelial Homeostasis. CURRENT STEM CELL REPORTS 2015. [DOI: 10.1007/s40778-014-0005-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
3184
|
Carding S, Verbeke K, Vipond DT, Corfe BM, Owen LJ. Dysbiosis of the gut microbiota in disease. MICROBIAL ECOLOGY IN HEALTH AND DISEASE 2015; 26:26191. [PMID: 25651997 PMCID: PMC4315779 DOI: 10.3402/mehd.v26.26191] [Citation(s) in RCA: 785] [Impact Index Per Article: 78.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
There is growing evidence that dysbiosis of the gut microbiota is associated with the pathogenesis of both intestinal and extra-intestinal disorders. Intestinal disorders include inflammatory bowel disease, irritable bowel syndrome (IBS), and coeliac disease, while extra-intestinal disorders include allergy, asthma, metabolic syndrome, cardiovascular disease, and obesity. In many of these conditions, the mechanisms leading to disease development involves the pivotal mutualistic relationship between the colonic microbiota, their metabolic products, and the host immune system. The establishment of a ‘healthy’ relationship early in life appears to be critical to maintaining intestinal homeostasis. Whilst we do not yet have a clear understanding of what constitutes a ‘healthy’ colonic microbiota, a picture is emerging from many recent studies identifying particular bacterial species associated with a healthy microbiota. In particular, the bacterial species residing within the mucus layer of the colon, either through direct contact with host cells, or through indirect communication via bacterial metabolites, may influence whether host cellular homeostasis is maintained or whether inflammatory mechanisms are triggered. In addition to inflammation, there is some evidence that perturbations in the gut microbiota is involved with the development of colorectal cancer. In this case, dysbiosis may not be the most important factor, rather the products of interaction between diet and the microbiome. High-protein diets are thought to result in the production of carcinogenic metabolites from the colonic microbiota that may result in the induction of neoplasia in the colonic epithelium. Ever more sensitive metabolomics methodologies reveal a suite of small molecules produced in the microbiome which mimic or act as neurosignallers or neurotransmitters. Coupled with evidence that probiotic interventions may alter psychological endpoints in both humans and in rodent models, these data suggest that CNS-related co-morbidities frequently associated with GI disease may originate in the intestine as a result of microbial dysbiosis. This review outlines the current evidence showing the extent to which the gut microbiota contributes to the development of disease. Based on evidence to date, we can assess the potential to positively modulate the composition of the colonic microbiota and ameliorate disease activity through bacterial intervention.
Collapse
Affiliation(s)
- Simon Carding
- Institute of Food Research, Norwich, UK.,Norwich Medical School, University of East Anglia, Norwich, UK
| | - Kristin Verbeke
- Translational Research in GastroIntestinal Disorders, KU Leuven, Leuven, Belgium
| | - Daniel T Vipond
- Institute of Food Research, Norwich, UK.,Norwich Medical School, University of East Anglia, Norwich, UK
| | - Bernard M Corfe
- Molecular Gastroenterology Research Group, Department of Oncology, University of Sheffield, Sheffield, UK.,Insigneo Institute for in silico Medicine, University of Sheffield, Sheffield, UK;
| | - Lauren J Owen
- Human Nutrition Unit, Department of Oncology, University of Sheffield, Sheffield, UK
| |
Collapse
|
3185
|
Affiliation(s)
- Jerome Boursier
- Service d’Hépato-Gastroentérologie, Centre Hospitalier Universitaire d’Angers, Angers, France
- HIFIH, UPRES 3859, SFR 4208, Université LUNAM, Angers, France
| | - Anna Mae Diehl
- Division of Gastroenterology, Department of Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
3186
|
Carding S, Verbeke K, Vipond DT, Corfe BM, Owen LJ. Dysbiosis of the gut microbiota in disease. MICROBIAL ECOLOGY IN HEALTH AND DISEASE 2015. [PMID: 25651997 DOI: 10.3402/mehd.v3426.26191] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
There is growing evidence that dysbiosis of the gut microbiota is associated with the pathogenesis of both intestinal and extra-intestinal disorders. Intestinal disorders include inflammatory bowel disease, irritable bowel syndrome (IBS), and coeliac disease, while extra-intestinal disorders include allergy, asthma, metabolic syndrome, cardiovascular disease, and obesity.
Collapse
Affiliation(s)
- Simon Carding
- Institute of Food Research, Norwich, UK
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - Kristin Verbeke
- Translational Research in GastroIntestinal Disorders, KU Leuven, Leuven, Belgium
| | - Daniel T Vipond
- Institute of Food Research, Norwich, UK
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - Bernard M Corfe
- Molecular Gastroenterology Research Group, Department of Oncology, University of Sheffield, Sheffield, UK
- Insigneo Institute for in silico Medicine, University of Sheffield, Sheffield, UK;
| | - Lauren J Owen
- Human Nutrition Unit, Department of Oncology, University of Sheffield, Sheffield, UK
| |
Collapse
|
3187
|
Moroney NC, O'Grady MN, Robertson RC, Stanton C, O'Doherty JV, Kerry JP. Influence of level and duration of feeding polysaccharide (laminarin and fucoidan) extracts from brown seaweed (Laminaria digitata) on quality indices of fresh pork. Meat Sci 2015; 99:132-41. [PMID: 25443973 DOI: 10.1016/j.meatsci.2014.08.016] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Revised: 08/19/2014] [Accepted: 08/28/2014] [Indexed: 11/20/2022]
Abstract
The effect of level (450 or 900 mg laminarin (L) and fucoidan (F) /kg feed) and duration (3 or 6 wks) of feeding a seaweed (Laminaria digitata) extract containing L/F on the quality of pork (longissimus thoracis et lumborum (LTL)) stored in modified atmosphere packs and on organ lipid stability was examined. Mechanisms of L/F antioxidant activity in LTL were evaluated. Plasma total antioxidant status, LTL pH, colour, microbiology and 'eating quality' sensory analysis were unaffected by dietary L/F. 'Visual' sensory descriptors (purchasing appeal and overall visual acceptability) were enhanced (p<0.05) in L/F450-3 LTL. Lipid oxidation was lower (p<0.05) in L/F450-3 and L/F900-3 LTL and reduced in L/F900-6 kidney homogenates. In cooked minced pork, lipid oxidation was not reduced by dietary L/F. Saturated fatty acids were lower (p<0.05) in L/F900-6 LTL. Results indicated L/F in pig diets for 3 weeks enhanced pork quality.
Collapse
Affiliation(s)
- N C Moroney
- Food Packaging Group, School of Food and Nutritional Sciences, College of Science, Engineering and Food Science, University College Cork, Ireland
| | - M N O'Grady
- Food Packaging Group, School of Food and Nutritional Sciences, College of Science, Engineering and Food Science, University College Cork, Ireland
| | - R C Robertson
- Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork, Ireland; School of Microbiology, University College Cork, Co. Cork, Ireland
| | - C Stanton
- Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork, Ireland; Alimentary Pharmabiotic Centre, Cork, Ireland
| | - J V O'Doherty
- School of Agriculture, Food Science, and Veterinary Medicine, College of Life Sciences, Lyons Research Farm, University College Dublin, Newcastle, Co. Dublin, Ireland
| | - J P Kerry
- Food Packaging Group, School of Food and Nutritional Sciences, College of Science, Engineering and Food Science, University College Cork, Ireland.
| |
Collapse
|
3188
|
In situ prebiotics for weaning piglets: in vitro production and fermentation of potato galacto-rhamnogalacturonan. Appl Environ Microbiol 2014; 81:1668-78. [PMID: 25527557 DOI: 10.1128/aem.03582-14] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Postweaning diarrhea (PWD) in pigs is a leading cause of economic loss in pork production worldwide. The current practice of using antibiotics and zinc to treat PWD is unsustainable due to the potential of antibiotic resistance and ecological disturbance, and novel methods are required. In this study, an in vitro model was used to test the possibility of producing prebiotic fiber in situ in the gastrointestinal (GI) tract of the piglet and the prebiotic activity of the resulting fiber in the terminal ileum. Soluble fiber was successfully produced from potato pulp, an industrial waste product, with the minimal enzyme dose in a simulated upper GI tract model extracting 26.9% of the initial dry matter. The fiber was rich in galactose and galacturonic acid and was fermented at 2.5, 5, or 10 g/liter in a glucose-free medium inoculated with the gut contents of piglet terminal ileum. Fermentations of 5 g/liter inulin or 5 g/liter of a purified potato fiber were used as controls. The fibers showed high fermentability, evident by a dose-dependent drop in pH and an increase in the organic acid content, with lactate in particular being increased. Deep sequencing showed a significant increase in the numbers of Lactobacillus and Veillonella organisms and an insignificant increase in the numbers of Clostridium organisms as well as a decrease in the numbers of Streptococcus organisms. Multivariate analysis showed clustering of the treatment groups, with the group treated with purified potato fiber being clearly separated from the other groups, as the microbiota composition was 60% Lactobacillus and almost free of Clostridium. For animal studies, a dosage corresponding to the 5-g/liter treatment is suggested.
Collapse
|
3189
|
Kim S, Kim JH, Park BO, Kwak YS. Perspectives on the therapeutic potential of short-chain fatty acid receptors. BMB Rep 2014; 47:173-8. [PMID: 24499669 PMCID: PMC4163876 DOI: 10.5483/bmbrep.2014.47.3.272] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Revised: 12/19/2013] [Accepted: 01/13/2014] [Indexed: 01/14/2023] Open
Abstract
There is rapidly growing interest in the human microbiome because of its implication in metabolic disorders and inflammatory diseases. Consequently, understanding the biology of short chain fatty acids and their receptors has become very important for identifying novel therapeutic avenues. GPR41 and GPR43 have been recognized as the cognate receptors for SCFAs and their roles in metabolism and inflammation have drawn much attention in recent years. GPR43 is highly expressed on immune cells and has been suggested to play a role in inflammatory diseases such as inflammatory bowel disease. Both GPR41 and GPR43 have been implicated in diabetes and obesity via the regulation of adipose tissue and gastrointestinal hormones. So far, many studies have provided contradictory results, and therefore further research is required to validate these receptors as drug targets. We will also discuss the synthetic modulators of GPR41 and GPR43 that are critical to understanding the functions of these receptors. [BMB Reports 2014; 47(3): 173-178]
Collapse
Affiliation(s)
- Sunhong Kim
- Targeted Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Ochang, Chungbuk 363-883; Department of Biomolecular Science, University of Science and Technology, Daejeon 305-350, Korea
| | - Jeong-Hoon Kim
- Medical Proteomics Research Center, Korea Research Institute of Bioscience and Biotechnology, Ochang, Chungbuk 363-883; Department of Functional Genomics, University of Science and Technology, Daejeon 305-350, Korea
| | - Bi Oh Park
- Targeted Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Ochang, Chungbuk 363-883; College of Pharmacy, Chungbuk National University, Cheongju 361-763, Korea
| | - Young Shin Kwak
- College of Pharmacy, Korea University, Sejong 339-700, Korea
| |
Collapse
|
3190
|
Ha CWY, Lam YY, Holmes AJ. Mechanistic links between gut microbial community dynamics, microbial functions and metabolic health. World J Gastroenterol 2014; 20:16498-16517. [PMID: 25469018 PMCID: PMC4248193 DOI: 10.3748/wjg.v20.i44.16498] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2014] [Revised: 06/26/2014] [Accepted: 08/28/2014] [Indexed: 02/06/2023] Open
Abstract
Gut microbes comprise a high density, biologically active community that lies at the interface of an animal with its nutritional environment. Consequently their activity profoundly influences many aspects of the physiology and metabolism of the host animal. A range of microbial structural components and metabolites directly interact with host intestinal cells and tissues to influence nutrient uptake and epithelial health. Endocrine, neuronal and lymphoid cells in the gut also integrate signals from these microbial factors to influence systemic responses. Dysregulation of these host-microbe interactions is now recognised as a major risk factor in the development of metabolic dysfunction. This is a two-way process and understanding the factors that tip host-microbiome homeostasis over to dysbiosis requires greater appreciation of the host feedbacks that contribute to regulation of microbial community composition. To date, numerous studies have employed taxonomic profiling approaches to explore the links between microbial composition and host outcomes (especially obesity and its comorbidities), but inconsistent host-microbe associations have been reported. Available data indicates multiple factors have contributed to discrepancies between studies. These include the high level of functional redundancy in host-microbiome interactions combined with individual variation in microbiome composition; differences in study design, diet composition and host system between studies; and inherent limitations to the resolution of rRNA-based community profiling. Accounting for these factors allows for recognition of the common microbial and host factors driving community composition and development of dysbiosis on high fat diets. New therapeutic intervention options are now emerging.
Collapse
|
3191
|
Consolandi C, Turroni S, Emmi G, Severgnini M, Fiori J, Peano C, Biagi E, Grassi A, Rampelli S, Silvestri E, Centanni M, Cianchi F, Gotti R, Emmi L, Brigidi P, Bizzaro N, De Bellis G, Prisco D, Candela M, D'Elios MM. Behçet's syndrome patients exhibit specific microbiome signature. Autoimmun Rev 2014; 14:269-76. [PMID: 25435420 DOI: 10.1016/j.autrev.2014.11.009] [Citation(s) in RCA: 191] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 11/15/2014] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND AIMS Behçet syndrome is a systemic inflammatory condition characterized by muco-cutaneous and ocular manifestations, with central nervous system, vascular and/or gastro-intestinal involvement. The association of microbiota with Behçet syndrome has not been shown yet. Our work was aimed to compare the gut microbiota structure and the profiles of short-chain fatty acids production in Behçet syndrome patients and healthy control relatives. METHODS Here, we compared the fecal microbiota of 22 patients with Behçet syndrome and that of 16 healthy co-habiting controls, sharing the same diet and lifestyle by pyrosequencing of the V3-V4 hypervariable regions of the 16 rDNA gene and biochemical analyses. RESULTS Our analyses showed significant differences in gut microbiota between Behçet patients and healthy cohabitants. In particular we found that Behçet's patients were significantly depleted in the genera Roseburia and Subdoligranulum. Roseburia showed a relative abundance value of 10.45±6.01% in healthy relatives and 4.97±5.09% in Behçet's patients, and Subdoligranulum, which reached a relative abundance of 3.28±2.20% in healthy controls, was only at 1.93±1.75% of abundance in Behçet's patients. Here we report, for the first time, that a peculiar dysbiosis of the gut microbiota is present in patients with Behçet syndrome and this corresponds to specific changes in microbiome profile. A significant decrease of butyrate production (P=0.0033) in Behçet's patients was demonstrated. Butyrate is able to promote differentiation of T-regulatory cells, and consequently the results obtained prompt us to speculate that a defect of butyrate production might lead to both reduced T-reg responses and activation of immuno-pathological T-effector responses. CONCLUSIONS Altogether, our results indicate that both a peculiar dysbiosis of the gut microbiota and a significant decrease of butyrate production are present in patients with Behçet syndrome.
Collapse
Affiliation(s)
- Clarissa Consolandi
- Institute of Biomedical Technologies, National Research Council (ITB-CNR), Segrate, Milan, Italy.
| | - Silvia Turroni
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Giacomo Emmi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Marco Severgnini
- Institute of Biomedical Technologies, National Research Council (ITB-CNR), Segrate, Milan, Italy
| | - Jessica Fiori
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Clelia Peano
- Institute of Biomedical Technologies, National Research Council (ITB-CNR), Segrate, Milan, Italy
| | - Elena Biagi
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Alessia Grassi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Simone Rampelli
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Elena Silvestri
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Manuela Centanni
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Fabio Cianchi
- Department of Surgery and Translational Medicine, University of Florence, Italy
| | - Roberto Gotti
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Lorenzo Emmi
- Medical Pathology, Center for Autoimmune Systemic Diseases, Behçet Center and Lupus Clinic, AOU Careggi, Florence, Italy.
| | - Patrizia Brigidi
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Nicola Bizzaro
- Laboratory of Clinical Pathology, Diagnostic Department, San Antonio Hospital, Tolmezzo, Italy
| | - Gianluca De Bellis
- Institute of Biomedical Technologies, National Research Council (ITB-CNR), Segrate, Milan, Italy
| | - Domenico Prisco
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy; Medical Pathology, Center for Autoimmune Systemic Diseases, Behçet Center and Lupus Clinic, AOU Careggi, Florence, Italy
| | - Marco Candela
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Mario M D'Elios
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy; Medical Pathology, Center for Autoimmune Systemic Diseases, Behçet Center and Lupus Clinic, AOU Careggi, Florence, Italy
| |
Collapse
|
3192
|
Han J, Lin K, Sequeira C, Borchers CH. An isotope-labeled chemical derivatization method for the quantitation of short-chain fatty acids in human feces by liquid chromatography-tandem mass spectrometry. Anal Chim Acta 2014; 854:86-94. [PMID: 25479871 DOI: 10.1016/j.aca.2014.11.015] [Citation(s) in RCA: 375] [Impact Index Per Article: 34.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Revised: 11/05/2014] [Accepted: 11/13/2014] [Indexed: 12/19/2022]
Abstract
Short-chain fatty acids (SCFAs) are produced by anaerobic gut microbiota in the large bowel. Qualitative and quantitative measurements of SCFAs in the intestinal tract and the fecal samples are important to understand the complex interplay between diet, gut microbiota and host metabolism homeostasis. To develop a new LC-MS/MS method for sensitive and reliable analysis of SCFAs in human fecal samples, 3-nitrophenylhydrazine (3NPH) was employed for pre-analytical derivatization to convert ten C2-C6 SCFAs to their 3-nitrophenylhydrazones under a single set of optimized reaction conditions and without the need of reaction quenching. The derivatives showed excellent in-solution chemical stability. They were separated on a reversed-phase C18 column and quantitated by negative-ion electrospray ionization - multiple-reaction monitoring (MRM)/MS. To achieve accurate quantitation, the stable isotope-labeled versions of the derivatives were synthesized in a single reaction vessel from (13)C6-3NPH, and were used as internal standard to compensate for the matrix effects in ESI. Method validation showed on-column limits of detection and quantitation over the range from low to high femtomoles for the ten SCFAs, and the intra-day and inter-day precision for determination of nine of the ten SCFAs in human fecal samples was ≤8.8% (n=6). The quantitation accuracy ranged from 93.1% to 108.4% (CVs≤4.6%, n=6). This method was used to determine the SCFA concentrations and compositions in six human fecal samples. One of the six samples, which was collected from a clinically diagnosed type 2 diabetes patient showed a significantly high molar ratio of branch-chain SCFAs to straight-chain SCFAs than the others. In summary, this work provides a new LC-MS/MS method for precise and accurate quantitation of SCFAs in human feces.
Collapse
Affiliation(s)
- Jun Han
- University of Victoria - Genome BC Proteomics Centre, University of Victoria, Vancouver Island Technology Park, 3101-4464 Markham Street, Victoria, BC V8Z 7X8, Canada
| | - Karen Lin
- University of Victoria - Genome BC Proteomics Centre, University of Victoria, Vancouver Island Technology Park, 3101-4464 Markham Street, Victoria, BC V8Z 7X8, Canada
| | - Carita Sequeira
- University of Victoria - Genome BC Proteomics Centre, University of Victoria, Vancouver Island Technology Park, 3101-4464 Markham Street, Victoria, BC V8Z 7X8, Canada
| | - Christoph H Borchers
- University of Victoria - Genome BC Proteomics Centre, University of Victoria, Vancouver Island Technology Park, 3101-4464 Markham Street, Victoria, BC V8Z 7X8, Canada; Department of Biochemistry and Microbiology, University of Victoria, Petch Building Room 207, 3800 Finnerty Road, Victoria, BC V8P 5C2, Canada.
| |
Collapse
|
3193
|
Sharon G, Garg N, Debelius J, Knight R, Dorrestein PC, Mazmanian SK. Specialized metabolites from the microbiome in health and disease. Cell Metab 2014; 20:719-730. [PMID: 25440054 PMCID: PMC4337795 DOI: 10.1016/j.cmet.2014.10.016] [Citation(s) in RCA: 417] [Impact Index Per Article: 37.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The microbiota, and the genes that comprise its microbiome, play key roles in human health. Host-microbe interactions affect immunity, metabolism, development, and behavior, and dysbiosis of gut bacteria contributes to disease. Despite advances in correlating changes in the microbiota with various conditions, specific mechanisms of host-microbiota signaling remain largely elusive. We discuss the synthesis of microbial metabolites, their absorption, and potential physiological effects on the host. We propose that the effects of specialized metabolites may explain present knowledge gaps in linking the gut microbiota to biological host mechanisms during initial colonization, and in health and disease.
Collapse
Affiliation(s)
- Gil Sharon
- Division of Biology and Biological Engineering, California institute of Technology, Pasadena, CA 91125, USA
| | - Neha Garg
- Skaggs School of Pharmacy & Pharmaceutical Sciences, University of California at San Diego, La Jolla, CA 92093, USA
| | - Justine Debelius
- Department of Chemistry and Biochemistry, University of Colorado, Boulder, CO 80309, USA
| | - Rob Knight
- Department of Chemistry and Biochemistry, University of Colorado, Boulder, CO 80309, USA; Howard Hughes Medical Institute, Boulder, CO 80309, USA
| | - Pieter C Dorrestein
- Skaggs School of Pharmacy & Pharmaceutical Sciences, University of California at San Diego, La Jolla, CA 92093, USA; Department of Pharmacology, University of California at San Diego, La Jolla, CA 92093, USA
| | - Sarkis K Mazmanian
- Division of Biology and Biological Engineering, California institute of Technology, Pasadena, CA 91125, USA.
| |
Collapse
|
3194
|
den Besten G, Havinga R, Bleeker A, Rao S, Gerding A, van Eunen K, Groen AK, Reijngoud DJ, Bakker BM. The short-chain fatty acid uptake fluxes by mice on a guar gum supplemented diet associate with amelioration of major biomarkers of the metabolic syndrome. PLoS One 2014; 9:e107392. [PMID: 25203112 PMCID: PMC4159349 DOI: 10.1371/journal.pone.0107392] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Accepted: 08/15/2014] [Indexed: 01/13/2023] Open
Abstract
Studies with dietary supplementation of various types of fibers have shown beneficial effects on symptoms of the metabolic syndrome. Short-chain fatty acids (SCFAs), the main products of intestinal bacterial fermentation of dietary fiber, have been suggested to play a key role. Whether the concentration of SCFAs or their metabolism drives these beneficial effects is not yet clear. In this study we investigated the SCFA concentrations and in vivo host uptake fluxes in the absence or presence of the dietary fiber guar gum. C57Bl/6J mice were fed a high-fat diet supplemented with 0%, 5%, 7.5% or 10% of the fiber guar gum. To determine the effect on SCFA metabolism, 13C-labeled acetate, propionate or butyrate were infused into the cecum of mice for 6 h and the isotopic enrichment of cecal SCFAs was measured. The in vivo production, uptake and bacterial interconversion of acetate, propionate and butyrate were calculated by combining the data from the three infusion experiments in a single steady-state isotope model. Guar gum treatment decreased markers of the metabolic syndrome (body weight, adipose weight, triglycerides, glucose and insulin levels and HOMA-IR) in a dose-dependent manner. In addition, hepatic mRNA expression of genes involved in gluconeogenesis and fatty acid synthesis decreased dose-dependently by guar gum treatment. Cecal SCFA concentrations were increased compared to the control group, but no differences were observed between the different guar gum doses. Thus, no significant correlation was found between cecal SCFA concentrations and metabolic markers. In contrast, in vivo SCFA uptake fluxes by the host correlated linearly with metabolic markers. We argue that in vivo SCFA fluxes, and not concentrations, govern the protection from the metabolic syndrome by dietary fibers.
Collapse
Affiliation(s)
- Gijs den Besten
- Center for Liver, Digestive and Metabolic Diseases, Department of Pediatrics & Systems Biology Center for Energy Metabolism and Ageing, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Netherlands Consortium for Systems Biology, Amsterdam, The Netherlands
| | - Rick Havinga
- Center for Liver, Digestive and Metabolic Diseases, Department of Pediatrics & Systems Biology Center for Energy Metabolism and Ageing, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Aycha Bleeker
- Center for Liver, Digestive and Metabolic Diseases, Department of Pediatrics & Systems Biology Center for Energy Metabolism and Ageing, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Top Institute Food and Nutrition, Wageningen, The Netherlands
| | - Shodhan Rao
- Center for Liver, Digestive and Metabolic Diseases, Department of Pediatrics & Systems Biology Center for Energy Metabolism and Ageing, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Albert Gerding
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Karen van Eunen
- Center for Liver, Digestive and Metabolic Diseases, Department of Pediatrics & Systems Biology Center for Energy Metabolism and Ageing, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Netherlands Consortium for Systems Biology, Amsterdam, The Netherlands
- Top Institute Food and Nutrition, Wageningen, The Netherlands
| | - Albert K. Groen
- Center for Liver, Digestive and Metabolic Diseases, Department of Pediatrics & Systems Biology Center for Energy Metabolism and Ageing, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Netherlands Consortium for Systems Biology, Amsterdam, The Netherlands
| | - Dirk-Jan Reijngoud
- Center for Liver, Digestive and Metabolic Diseases, Department of Pediatrics & Systems Biology Center for Energy Metabolism and Ageing, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Netherlands Consortium for Systems Biology, Amsterdam, The Netherlands
| | - Barbara M. Bakker
- Center for Liver, Digestive and Metabolic Diseases, Department of Pediatrics & Systems Biology Center for Energy Metabolism and Ageing, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Netherlands Consortium for Systems Biology, Amsterdam, The Netherlands
- * E-mail:
| |
Collapse
|
3195
|
Abstract
A healthy gut microbiota plays many crucial functions in the host, being involved in the correct development and functioning of the immune system, assisting in the digestion of certain foods and in the production of health-beneficial bioactive metabolites or 'pharmabiotics'. These include bioactive lipids (including SCFA and conjugated linoleic acid) antimicrobials and exopolysaccharides in addition to nutrients, including vitamins B and K. Alterations in the composition of the gut microbiota and reductions in microbial diversity are highlighted in many disease states, possibly rendering the host susceptible to infection and consequently negatively affecting innate immune function. Evidence is also emerging of microbially produced molecules with neuroactive functions that can have influences across the brain-gut axis. For example, γ-aminobutyric acid, serotonin, catecholamines and acetylcholine may modulate neural signalling within the enteric nervous system, when released in the intestinal lumen and consequently signal brain function and behaviour. Dietary supplementation with probiotics and prebiotics are the most widely used dietary adjuncts to modulate the gut microbiota. Furthermore, evidence is emerging of the interactions between administered microbes and dietary substrates, leading to the production of pharmabiotics, which may directly or indirectly positively influence human health.
Collapse
|
3196
|
Psichas A, Sleeth ML, Murphy KG, Brooks L, Bewick GA, Hanyaloglu AC, Ghatei MA, Bloom SR, Frost G. The short chain fatty acid propionate stimulates GLP-1 and PYY secretion via free fatty acid receptor 2 in rodents. Int J Obes (Lond) 2014; 39:424-9. [PMID: 25109781 PMCID: PMC4356745 DOI: 10.1038/ijo.2014.153] [Citation(s) in RCA: 560] [Impact Index Per Article: 50.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Revised: 06/03/2014] [Accepted: 06/25/2014] [Indexed: 12/24/2022]
Abstract
Background and Objectives: The gut hormones peptide YY (PYY) and glucagon-like peptide 1 (GLP-1) acutely suppress appetite. The short chain fatty acid (SCFA) receptor, free fatty acid receptor 2 (FFA2) is present on colonic enteroendocrine L cells, and a role has been suggested for SCFAs in appetite regulation. Here, we characterise the in vitro and in vivo effects of colonic propionate on PYY and GLP-1 release in rodents, and investigate the role of FFA2 in mediating these effects using FFA2 knockout mice. Methods: We used Wistar rats, C57BL6 mice and free fatty acid receptor 2 knockout (FFA−/−) mice on a C57BL6 background to explore the impact of the SCFA propionate on PYY and GLP-1 release. Isolated colonic crypt cultures were used to assess the effects of propionate on gut hormone release in vitro. We subsequently developed an in vivo technique to assess gut hormone release into the portal vein following colonic infusion of propionate. Results: Propionate stimulated the secretion of both PYY and GLP-1 from wild-type primary murine colonic crypt cultures. This effect was significantly attenuated in cultures from FFA2−/− mice. Intra-colonic infusion of propionate elevated PYY and GLP-1 levels in jugular vein plasma in rats and in portal vein plasma in both rats and mice. However, propionate did not significantly stimulate gut hormone release in FFA2−/− mice. Conclusions: Intra-colonic administration of propionate stimulates the concurrent release of both GLP-1 and PYY in rats and mice. These data demonstrate that FFA2 deficiency impairs SCFA-induced gut hormone secretion both in vitro and in vivo.
Collapse
Affiliation(s)
- A Psichas
- Nutrition and Dietetic Research Group, Section of Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Imperial College, London, UK
| | - M L Sleeth
- Nutrition and Dietetic Research Group, Section of Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Imperial College, London, UK
| | - K G Murphy
- Section of Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Imperial College, London, UK
| | - L Brooks
- Section of Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Imperial College, London, UK
| | - G A Bewick
- 1] Section of Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Imperial College, London, UK [2] Division of Diabetes & Nutritional Sciences, Kings College London, Yeovil, UK
| | - A C Hanyaloglu
- Department of Surgery and Cancer, Institute of Reproductive and Developmental Biology, Imperial College, London, UK
| | - M A Ghatei
- Section of Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Imperial College, London, UK
| | - S R Bloom
- Section of Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Imperial College, London, UK
| | - G Frost
- Nutrition and Dietetic Research Group, Section of Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Imperial College, London, UK
| |
Collapse
|
3197
|
Running CA, Mattes RD. Different oral sensitivities to and sensations of short-, medium-, and long-chain fatty acids in humans. Am J Physiol Gastrointest Liver Physiol 2014; 307:G381-9. [PMID: 24924750 DOI: 10.1152/ajpgi.00181.2014] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Fatty acids that vary in chain length and degree of unsaturation have different effects on metabolism and human health. As evidence for a "taste" of nonesterified fatty acids (NEFA) accumulates, it may be hypothesized that fatty acid structures will also influence oral sensations. The present study examined oral sensitivity to caproic (C6), lauric (C12), and oleic (C18:1) acids over repeated visits. Analyses were also conducted on textural properties of NEFA emulsions and blank solutions. Oral thresholds for caproic acid were lower compared with oleic acid. Lauric acid thresholds were intermediate but not significantly different from either, likely due to lingering irritating sensations that prevented accurate discrimination. From particle size analysis, larger droplets were observed in blank solutions when mineral oil was used, leading to instability of the emulsion, which was not observed when emulsions contained NEFA or when mineral oil was removed from the blank. Rheological data showed no differences in viscosity among samples except for a slightly higher viscosity with oleic acid concentrations above 58 mM. Thus, texture was unlikely to be the property used to distinguish between the samples. Differences in oral detection and sensation of caproic, lauric, and oleic acids may be due to different properties of the fatty acid alkyl chains.
Collapse
Affiliation(s)
- Cordelia A Running
- Department of Food Science, Purdue University, West Lafayette, Indiana; and
| | - Richard D Mattes
- Department of Nutrition Science, Purdue University, West Lafayette, Indiana
| |
Collapse
|
3198
|
Rebello CJ, Greenway FL, Finley JW. Whole grains and pulses: a comparison of the nutritional and health benefits. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2014; 62:7029-7049. [PMID: 24992700 DOI: 10.1021/jf500932z] [Citation(s) in RCA: 143] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Nutrition plays an important role in the prevention and management of disease. Whole grain cereals contain a host of nutrients and bioactive substances that have health-promoting effects. Epidemiological evidence shows a consistent inverse association between whole grain intake and the risk of chronic disease. Despite a concerted effort by scientists, educators, and policy makers to promote the consumption of whole grains, it remains dismally short of the recommended intakes. Pulses (dried beans and peas) differ from whole grains in their structural and physicochemical properties and have varying amounts of fiber, resistant starch, vitamins, minerals, and other bioactive components; nevertheless, these food groups complement each other. Observational as well as intervention trials show that pulse consumption has beneficial effects on the prevention and management of chronic disease. The nutritional and phytochemical components of pulses coupled with those of whole grains suggest a potential synergistic effect that could provide significant health benefits.
Collapse
Affiliation(s)
- Candida J Rebello
- School of Nutrition and Food Sciences, Louisiana State University , Baton Rouge, Louisiana 70803, United States
| | | | | |
Collapse
|
3199
|
Eberle JAM, Widmayer P, Breer H. Receptors for short-chain fatty acids in brush cells at the "gastric groove". Front Physiol 2014; 5:152. [PMID: 24795647 PMCID: PMC3997014 DOI: 10.3389/fphys.2014.00152] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Accepted: 03/31/2014] [Indexed: 01/21/2023] Open
Abstract
In the stomach of rodents clusters of brush cells are arranged at the "gastric groove," immediately at the transition zone from the non-glandular reservoir compartment to the glandular digestive compartment. Based on their taste cell-like molecular phenotype it has been speculated that the cells may be capable to sense constituents of the ingested food, however, searches for nutrient receptors have not been successful. In this study, it was hypothesized that the cells may express receptors for short-chain fatty acids, metabolites generated by microorganisms during the storage of ingested food in the murine forestomach, which lacks the acidic milieu of more posterior regions of the stomach and is colonized with numerous microbiota. Experimental approaches, including RT-PCR analysis and immunohistochemical studies, revealed that the majority of these brush cells express the G-protein coupled receptor types GPR41 (FFAR3) and GPR43 (FFAR2), which are activated by short-chain fatty acids. Both, the GPR41 receptor proteins as well as an appropriate G-protein, α-gustducin, were found to be segregated at the apical brush border of the cells, indicating a direct contact with the luminal content of this gastric region. The exposure of microvillar processes with appropriate receptors and signaling elements to the gastric lumen suggests that the brush cells may in fact be capable to sense the short-chain fatty acids which originate from fermentation processes during the retention of ingested food in the anterior part of the stomach.
Collapse
Affiliation(s)
| | | | - Heinz Breer
- Institute of Physiology, University of Hohenheim Stuttgart, Germany
| |
Collapse
|
3200
|
Schnorr SL, Candela M, Rampelli S, Centanni M, Consolandi C, Basaglia G, Turroni S, Biagi E, Peano C, Severgnini M, Fiori J, Gotti R, De Bellis G, Luiselli D, Brigidi P, Mabulla A, Marlowe F, Henry AG, Crittenden AN. Gut microbiome of the Hadza hunter-gatherers. Nat Commun 2014; 5:3654. [PMID: 24736369 PMCID: PMC3996546 DOI: 10.1038/ncomms4654] [Citation(s) in RCA: 854] [Impact Index Per Article: 77.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Accepted: 03/14/2014] [Indexed: 02/06/2023] Open
Abstract
Human gut microbiota directly influences health and provides an extra means of adaptive potential to different lifestyles. To explore variation in gut microbiota and to understand how these bacteria may have co-evolved with humans, here we investigate the phylogenetic diversity and metabolite production of the gut microbiota from a community of human hunter-gatherers, the Hadza of Tanzania. We show that the Hadza have higher levels of microbial richness and biodiversity than Italian urban controls. Further comparisons with two rural farming African groups illustrate other features unique to Hadza that can be linked to a foraging lifestyle. These include absence of Bifidobacterium and differences in microbial composition between the sexes that probably reflect sexual division of labour. Furthermore, enrichment in Prevotella, Treponema and unclassified Bacteroidetes, as well as a peculiar arrangement of Clostridiales taxa, may enhance the Hadza’s ability to digest and extract valuable nutrition from fibrous plant foods. Gut microbes influence our health and may contribute to human adaptation to different lifestyles. Here, the authors describe the gut microbiome of a community of hunter-gatherers and identify unique features that could be linked to a foraging lifestyle.
Collapse
Affiliation(s)
- Stephanie L Schnorr
- 1] Plant Foods in Hominin Dietary Ecology Research Group, Max Planck Institute for Evolutionary Anthropology, Deutscher Platz 6, 04103 Leipzig, Germany [2]
| | - Marco Candela
- 1] Department of Pharmacy and Biotechnology, University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy [2]
| | - Simone Rampelli
- Department of Pharmacy and Biotechnology, University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Manuela Centanni
- Department of Pharmacy and Biotechnology, University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Clarissa Consolandi
- Institute of Biomedical Technologies, Italian National Research Council, Via Fratelli Cervi 93, 20090 Segrate, Milan, Italy
| | - Giulia Basaglia
- Department of Pharmacy and Biotechnology, University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Silvia Turroni
- Department of Pharmacy and Biotechnology, University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Elena Biagi
- Department of Pharmacy and Biotechnology, University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Clelia Peano
- Institute of Biomedical Technologies, Italian National Research Council, Via Fratelli Cervi 93, 20090 Segrate, Milan, Italy
| | - Marco Severgnini
- Institute of Biomedical Technologies, Italian National Research Council, Via Fratelli Cervi 93, 20090 Segrate, Milan, Italy
| | - Jessica Fiori
- Department of Pharmacy and Biotechnology, University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Roberto Gotti
- Department of Pharmacy and Biotechnology, University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Gianluca De Bellis
- Institute of Biomedical Technologies, Italian National Research Council, Via Fratelli Cervi 93, 20090 Segrate, Milan, Italy
| | - Donata Luiselli
- Department of Biological, Geological and Environmental Sciences, University of Bologna, Via Selmi 3, Bologna 40126, Italy
| | - Patrizia Brigidi
- Department of Pharmacy and Biotechnology, University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Audax Mabulla
- College of Arts and Social Sciences, University of Dar es Salaam, 35091 Dar es Salaam, Tanzania
| | - Frank Marlowe
- Division of Biological Anthropology, University of Cambridge, Cambridge CB2 1TN, UK
| | - Amanda G Henry
- Plant Foods in Hominin Dietary Ecology Research Group, Max Planck Institute for Evolutionary Anthropology, Deutscher Platz 6, 04103 Leipzig, Germany
| | - Alyssa N Crittenden
- Metabolism, Anthropometry, and Nutrition Laboratory, Department of Anthropology, University of Nevada, Las Vegas, Nevada 89154-5003, USA
| |
Collapse
|