Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 31
1.
Bioorg Med Chem Lett ; 107: 129758, 2024 Jul 15.
Article En | MEDLINE | ID: mdl-38641152

GPR41, a G protein-coupled receptor, serves as a sensor for short-chain fatty acids and plays a crucial role in regulating multiple physiological processes such as the maintenance of metabolic and immune homeostasis. Therefore, the modulation of GPR41 has garnered attention as a potential strategy for the treatment of various disorders. We conducted a structure-activity relationship study on a lead tetrahydroquinolone derivative bearing a 2-(trifluoromethoxy)benzene group that displayed antagonistic activity toward GPR41. Modification of the aryl group attached to the furan moiety revealed that derivatives containing di- or trifluorobenzene, instead of 2-(trifluoromethoxy)benzene, exhibited agonistic activity toward GPR41, comparable with the reported agonistic modulator AR420626. These results suggest that the aryl group plays a pivotal role in regulating the activity of compounds toward GPR41, providing valuable insights for the design of GPR41 modulators.


Receptors, G-Protein-Coupled , Structure-Activity Relationship , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/agonists , Humans , Molecular Structure , Quinolones/chemistry , Quinolones/pharmacology , Quinolones/chemical synthesis , Dose-Response Relationship, Drug , Receptors, Cell Surface
2.
Mol Nutr Food Res ; 68(9): e2300779, 2024 May.
Article En | MEDLINE | ID: mdl-38632845

The intestinal immune system plays a crucial role in obesity and insulin resistance. An altered intestinal immunity is associated with changes to the gut microbiota, barrier function, and tolerance to luminal antigens. Lipid metabolism and its unbalance can also contribute to acute and chronic inflammation in different conditions. In celiac disease (CD), the serum phospholipid profile in infants who developed CD is dramatically different when compared to that of infants at risk of CD not developing the disease. In a mouse model of gluten sensitivity, oral wheat gliadin challenge in connection with inhibition of the metabolism of arachidonic acid, an omega-6 polyunsaturated fatty acid, specifically induces the enteropathy. Recent evidence suggests that gluten may play a role also for development of life-style related diseases in populations on a high fat diet (HFD). However, the mechanisms behind these effects are not yet understood. Exploratory studies in mice feed HFD show that wheat gliadin consumption affects glucose and lipid metabolic homeostasis, alters the gut microbiota, and the immune cell profile in liver.


Celiac Disease , Diet, High-Fat , Gastrointestinal Microbiome , Gliadin , Obesity , Animals , Diet, High-Fat/adverse effects , Obesity/metabolism , Humans , Gastrointestinal Microbiome/physiology , Triticum , Mice , Lipid Metabolism
3.
Mol Nutr Food Res ; 68(9): e2300758, 2024 May.
Article En | MEDLINE | ID: mdl-38639319

SCOPE: Obesity and metabolic diseases are closely associated, and individuals who become obese are also prone to type 2 diabetes and cardiovascular disorders. Gut microbiota is mediated by diet and can influence host metabolism and the incidence of metabolic disorders. Recent studies have suggested that improving gut microbiota through a fructooligosaccharide (FOS)-supplemented diet may ameliorate obesity and other metabolic disorders. Although accumulating evidence supports the notion of the developmental origins of health and disease, the underlying mechanisms remain obscure. METHODS AND RESULTS: ICR mice are fed AIN-93G formula-based cellulose -, FOS-, acetate-, or propionate-supplemented diets during pregnancy. Offspring are reared by conventional ICR foster mothers for 4 weeks; weaned mice are fed high fat diet for 12 weeks and housed individually. The FOS and propionate offspring contribute to suppressing obesity and improving glucose intolerance. Gut microbial compositions in FOS-fed mothers and their offspring are markedly changed. However, the beneficial effect of FOS diet on the offspring is abolished when antibiotics are administered to pregnant mice. CONCLUSION: The findings highlight the link between the maternal gut environment and the developmental origin of metabolic syndrome in offspring. These results open novel research avenues into preemptive therapies for metabolic disorders by targeting the maternal gut microbiota.


Diet, High-Fat , Gastrointestinal Microbiome , Mice, Inbred ICR , Obesity , Oligosaccharides , Animals , Pregnancy , Oligosaccharides/pharmacology , Oligosaccharides/administration & dosage , Diet, High-Fat/adverse effects , Female , Gastrointestinal Microbiome/drug effects , Male , Mice, Obese , Mice , Prenatal Exposure Delayed Effects , Phenotype , Maternal Nutritional Physiological Phenomena , Dietary Supplements
4.
Anal Chem ; 96(3): 991-996, 2024 01 23.
Article En | MEDLINE | ID: mdl-38206184

Untargeted lipidomics using liquid chromatography (LC) coupled with tandem mass spectrometry (MS) is essential for large cohort studies. Using a fast LC gradient of less than 10 min for the rapid screening of lipids decreases the annotation rate, because of the lower coverage of the MS/MS spectra caused by the narrow peak width. A systematic procedure is proposed in this study to achieve a high annotation rate in fast LC-based untargeted lipidomics by integrating data-dependent acquisition (DDA) and sequential window acquisition of all-theoretical mass spectrometry data-independent acquisition (SWATH-DIA) techniques using the updated MS-DIAL program. This strategy uses variable SWATH-DIA methods for quality control (QC) samples, which are a mixture of biological samples that were analyzed multiple times to correct the MS signal drift. In contrast, biological samples are analyzed using DDA to facilitate the structural elucidation of lipids using the pure spectrum to the maximum extent. The workflow is demonstrated using an 8.6 min LC gradient, where the QC samples are analyzed using five different SWATH-DIA methods. The use of both DDA and SWATH-DIA achieves a 1.7-fold annotation coverage from publicly available benchmark data obtained using a fast LC-DDA-MS technique and offers 95.3% lipid coverage, as compared to the benchmark data set from a 25 min LC gradient. This study demonstrates that harmonized improvements in analytical conditions and informatics tools provide a comprehensive lipidome in fast LC-based untargeted lipidomics, not only for large-scale studies but also for small-scale experiments, contributing to both clinical applications and basic biology.


Lipidomics , Tandem Mass Spectrometry , Humans , Lipidomics/methods , Tandem Mass Spectrometry/methods , Chromatography, Liquid/methods , Liquid Chromatography-Mass Spectrometry , Lipids
5.
Obes Res Clin Pract ; 17(5): 411-420, 2023.
Article En | MEDLINE | ID: mdl-37679239

Orlistat, an anti-obesity agent, inhibits the metabolism and absorption of dietary fat by inactivating pancreatic lipase in the gut. The effect of orlistat on the gut microbiota of Japanese individuals with obesity is unknown. This study aimed to explore the effects of orlistat on the gut microbiota and fatty acid metabolism of Japanese individuals with obesity. Fourteen subjects with visceral fat obesity (waist circumference ≥85 cm) took orlistat orally at a dose of 60 mg, 3 times a day for 8 weeks. Body weight; waist circumference; visceral fat area; levels of short-chain fatty acids, gut microbiota, fatty acid metabolites in the feces, and gastrointestinal hormones; and adverse events were evaluated. Body weight, waist circumference, and blood leptin concentrations were significantly lower after orlistat treatment (mean ± standard deviation, 77.8 ± 9.1 kg; 91.9 ± 8.7 cm; and 4546 ± 3211 pg/mL, respectively) compared with before treatment (79.4 ± 9.0 kg; 94.4 ± 8.0 cm; and 5881 ± 3526 pg/mL, respectively). Significant increases in fecal levels of fatty acid metabolites (10-hydroxy-cis-12-octadecenoic acid, 10-oxo-cis-12-octadecenoic acid, and 10-oxo-trans-11-octadecenoic acid) were detected. Meanwhile, no significant changes were found in abdominal computed tomography parameters, blood marker levels, or short-chain fatty acid levels in the feces. Gut microbiota analysis revealed that some study subjects had decreased abundance of Firmicutes, increased abundance of Bacteroidetes, and increased α-diversity indices (Chao1 and ACE) after 8 weeks of treatment. The levels of Lactobacillus genus and Lactobacillus gasseri were significantly higher after 8 weeks of treatment. None of the subjects discontinued treatment or experienced severe adverse events. This study suggested that orlistat might alter gut microbiota composition and affect the body through fatty acid metabolites produced by the modified gut bacteria.


Gastrointestinal Microbiome , Humans , Orlistat/pharmacology , Obesity , Body Weight , Fatty Acids , Lipase
6.
Sci Rep ; 13(1): 5556, 2023 04 05.
Article En | MEDLINE | ID: mdl-37019935

Diet is the primary factor affecting host nutrition and metabolism, with excess food intake, especially high-calorie diets, such as high-fat and high-sugar diets, causing an increased risk of obesity and related disorders. Obesity alters the gut microbial composition and reduces microbial diversity and causes changes in specific bacterial taxa. Dietary lipids can alter the gut microbial composition in obese mice. However, the regulation of gut microbiota and host energy homeostasis by different polyunsaturated fatty acids (PUFAs) in dietary lipids remains unknown. Here, we demonstrated that different PUFAs in dietary lipids improved host metabolism in high-fat diet (HFD)-induced obesity in mice. The intake of the different PUFA-enriched dietary lipids improved metabolism in HFD-induced obesity by regulating glucose tolerance and inhibiting colonic inflammation. Moreover, the gut microbial compositions were different among HFD and modified PUFA-enriched HFD-fed mice. Thus, we have identified a new mechanism underlying the function of different PUFAs in dietary lipids in regulating host energy homeostasis in obese conditions. Our findings shed light on the prevention and treatment of metabolic disorders by targeting the gut microbiota.


Diet, High-Fat , Dietary Fats , Mice , Animals , Diet, High-Fat/adverse effects , Dietary Fats/pharmacology , Obesity/metabolism , Fatty Acids, Unsaturated/adverse effects , Inflammation/metabolism , Mice, Inbred C57BL , Lipid Metabolism
7.
J Oleo Sci ; 72(3): 303-312, 2023.
Article En | MEDLINE | ID: mdl-36878584

Dietary fish oil containing n-3 polyunsaturated fatty acids provides health benefits by lowering lipid levels in the liver and serum. ß-Conglycinin (ßCG) is a major constituent protein in soybean with many physiological effects, such as lowering blood triglyceride levels, preventing obesity and diabetes, and improving hepatic lipid metabolism. However, the combined effects of fish oil and ßCG remain unclear. Here, we investigated the effects of a dietary combination of fish oil and ßCG on lipid and glucose parameters in diabetic/obese KK-A y mice. KK-A y mice were divided into three groups: control, fish oil, and fish oil + ßCG; these groups were fed a casein-based diet containing 7% (w/w) soybean oil, a casein-based diet containing 2% (w/w) soybean oil and 5% (w/w) fish oil, and a ßCG-based diet containing 2% (w/w) soybean oil and 5% (w/w) fish oil, respectively. The effects of the dietary combination of fish oil and ßCG on blood biochemical parameters, adipose tissue weight, expression levels of fat- and glucose metabolism-related genes, and cecal microbiome composition were evaluated. The total white adipose tissue weight (p < 0.05), levels of total serum cholesterol (p < 0.01), triglyceride (p < 0.01), and blood glucose (p < 0.05), and expression levels of fatty acid synthesis-related genes (including Fasn (p < 0.05) and Acc (p < 0.05)), and glucose metabolism-related genes (such as Pepck (p < 0.05)) were lower in the fish oil and fish oil + ßCG groups than in the control group. Furthermore, the relative abundance of Bacteroidaceae and Coriobacteriaceae differed significantly between the fish oil + ßCG and control groups. These findings suggest that dietary intake of fish oil + ßCG may prevent obesity and diabetes, alleviate lipid abnormalities, and alter the gut microbiome composition in diabetic/obese KK-A y mice. Further research is needed to build on this study to evaluate the health benefits of major components of Japanese food.


Diabetes Mellitus , Fatty Acids, Omega-3 , Gastrointestinal Microbiome , Animals , Mice , Fish Oils/pharmacology , Blood Glucose , Caseins , Soybean Oil/pharmacology , Diet , Adipose Tissue , Fatty Acids, Omega-3/pharmacology , Obesity
8.
Int J Mol Sci ; 24(4)2023 Feb 14.
Article En | MEDLINE | ID: mdl-36835245

Inflammatory bowel disease (IBD) is an inflammatory disease of the gastrointestinal tract, the incidence of which has rapidly increased worldwide, especially in developing and Western countries. Recent research has suggested that genetic factors, the environment, microbiota, and immune responses are involved in the pathogenesis; however, the underlying causes of IBD are unclear. Recently, gut microbiota dysbiosis, especially a decrease in the abundance and diversity of specific genera, has been suggested as a trigger for IBD-initiating events. Improving the gut microbiota and identifying the specific bacterial species in IBD are essential for understanding the pathogenesis and treatment of IBD and autoimmune diseases. Here, we review the different aspects of the role played by gut microbiota in the pathogenesis of IBD and provide a theoretical basis for modulating gut microbiota through probiotics, fecal microbiota transplantation, and microbial metabolites.


Gastrointestinal Microbiome , Inflammatory Bowel Diseases , Microbiota , Probiotics , Humans , Bacteria , Fecal Microbiota Transplantation , Dysbiosis/microbiology
9.
Gut Microbes ; 15(1): 2161271, 2023.
Article En | MEDLINE | ID: mdl-36604628

Fermented foods demonstrate remarkable health benefits owing to probiotic bacteria or microproducts produced via bacterial fermentation. Fermented foods are produced by the fermentative action of several lactic acid bacteria, including Leuconostoc mesenteroides; however, the exact mechanism of action of these foods remains unclear. Here, we observed that prebiotics associated with L. mesenteroides-produced exopolysaccharides (EPS) demonstrate substantial host metabolic benefits. L. mesenteroides-produced EPS is an indigestible α-glucan, and intake of the purified form of EPS improved glucose metabolism and energy homeostasis through EPS-derived gut microbial short-chain fatty acids, and changed gut microbial composition. Our findings reveal an important mechanism that accounts for the effects of diet, prebiotics, and probiotics on energy homeostasis and suggests an approach for preventing lifestyle-related diseases by targeting bacterial EPS.


Gastrointestinal Microbiome , Lactobacillales , Leuconostoc mesenteroides , Probiotics , Prebiotics , Lactobacillales/metabolism , Bacteria , Fermentation
10.
Biochem Biophys Res Commun ; 621: 176-182, 2022 09 17.
Article En | MEDLINE | ID: mdl-35841764

We previously found that glucagon-like peptide 1 (GLP-1) secretion by co-administration of maltose plus an α-glucosidase inhibitor miglitol (maltose/miglitol) was suppressed by a GLUT2 inhibitor phloretin in mice. In addition, maltose/miglitol inhibited glucose-dependent insulinotropic polypeptide (GIP) secretion through a mechanism involving short chain fatty acids (SCFAs) produced by microbiome. However, it remains unknown whether phloretin suppresses GLP-1 secretion by modulating SCFAs. In this study, we examined the effect of phloretin on SCFA release from microbiome in vitro and in vivo. In Escherichia coli, acetate release into the medium was suppressed by phloretin, when cultured with maltose/miglitol. In mice, phloretin inhibited maltose/miglitol-induced SCFA increase in the portal vein. In addition, alpha methyl-d-glucose (αMDG), a poor substrate for GLUT2, significantly increased GLP-1 secretion when co-administered with phloridzin in mice, suggesting that GLUT2 is not essential for glucose/phloridzin-induced GLP-1 secretion. αMDG increased portal SCFA levels, thereby increasing GLP-1 secretion and suppressing GIP secretion in mice, suggesting that αMDG is metabolizable not for mammals, but for microbiota. In conclusion, phloretin is suggested to suppress maltose/miglitol-induced GLP-1 secretion via inhibiting SCFAs produced by microbiome.


Gastrointestinal Microbiome , Glucagon-Like Peptide 1 , Animals , Fatty Acids, Volatile , Gastric Inhibitory Polypeptide , Glucose , Maltose , Mammals , Mice , Phloretin/pharmacology , Phlorhizin , Receptors, G-Protein-Coupled
11.
Int J Mol Sci ; 23(10)2022 May 16.
Article En | MEDLINE | ID: mdl-35628369

Due to the excess energy intake, which is a result of a high fat and high carbohydrate diet, dysfunction of energy balance leads to metabolic disorders such as obesity and type II diabetes mellitus (T2DM). Since obesity can be a risk factor for various diseases, including T2DM, hypertension, hyperlipidemia, and metabolic syndrome, novel prevention and treatment are expected. Moreover, host diseases linked to metabolic disorders are associated with changes in gut microbiota profile. Gut microbiota is affected by diet, and nutrients are used as substrates by gut microbiota for produced metabolites, such as short-chain and long-chain fatty acids, that may modulate host energy homeostasis. These free fatty acids are not only essential energy sources but also signaling molecules via G-protein coupled receptors (GPCRs). Some GPCRs are critical for metabolic functions, such as hormone secretion and immune function in various types of cells and tissues and contribute to energy homeostasis. The current studies have shown that GPCRs for gut microbial metabolites improved host energy homeostasis and systemic metabolic disorders. Here, we will review the association between diet, gut microbiota, and host energy homeostasis.


Diet , Gastrointestinal Microbiome , Metabolic Diseases , Receptors, G-Protein-Coupled , Diabetes Mellitus, Type 2/complications , Gastrointestinal Microbiome/physiology , Homeostasis , Humans , Metabolic Diseases/metabolism , Obesity/metabolism , Receptors, G-Protein-Coupled/metabolism
12.
Int J Mol Sci ; 22(19)2021 Oct 06.
Article En | MEDLINE | ID: mdl-34639136

BACKGROUND: α-cyclodextrin (α-CD) is one of the dietary fibers that may have a beneficial effect on cholesterol and/or glucose metabolism, but its efficacy and mode of action remain unclear. METHODS: In the present study, we examined the anti-hyperglycemic effect of α-CD after oral loading of glucose and liquid meal in mice. RESULTS: Administration of 2 g/kg α-CD suppressed hyperglycemia after glucose loading, which was associated with increased glucagon-like peptide 1 (GLP-1) secretion and enhanced hepatic glucose sequestration. By contrast, 1 g/kg α-CD similarly suppressed hyperglycemia, but without increasing secretions of GLP-1 and insulin. Furthermore, oral α-CD administration disrupts lipid micelle formation through its inclusion of lecithin in the gut luminal fluid. Importantly, prior inclusion of α-CD with lecithin in vitro nullified the anti-hyperglycemic effect of α-CD in vivo, which was associated with increased intestinal mRNA expressions of SREBP2-target genes (Ldlr, Hmgcr, Pcsk9, and Srebp2). CONCLUSIONS: α-CD elicits its anti-hyperglycemic effect after glucose loading by inducing lecithin inclusion in the gut lumen and activating SREBP2, which is known to induce cholecystokinin secretion to suppress hepatic glucose production via a gut/brain/liver axis.


Gastrointestinal Tract/metabolism , Hyperglycemia/prevention & control , Lecithins/metabolism , Postprandial Period , Potassium Channels, Inwardly Rectifying/physiology , Sterol Regulatory Element Binding Protein 2/metabolism , alpha-Cyclodextrins/pharmacology , Animals , Gastrointestinal Tract/drug effects , Hyperglycemia/etiology , Hyperglycemia/metabolism , Hyperglycemia/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Sterol Regulatory Element Binding Protein 2/genetics
13.
Biochem Biophys Res Commun ; 557: 48-54, 2021 06 11.
Article En | MEDLINE | ID: mdl-33862459

Olfactory receptor 78 (Olfr78), which is also known as a receptor for short-chain fatty acids (SCFAs) produced via gut microbial fermentation from indigestible polysaccharides such as dietary fibers, is expressed in the enteroendocrine cells of the colon. However, the role of Olfr78 in gut hormone secretion remains unknown. Here, we aimed to investigate the function and mechanism of action of Olfr78 in vivo and in vitro. Toward this, we assessed the expression of Olfr78 in several tissues, affinity of Olfr78 to various monocarboxylates, and the secretion of anorexigenic gut hormone peptide YY (PYY) via Olfr78 using various molecular and biochemical techniques. Olfr78 was abundantly expressed in the colon and mouse enteroendocrine cell line STC-1 and showed specific affinity to SCFAs such as acetate and propionate, but not butyrate, in a monocarboxylate ligand screening assay using a heterologous expression system. Acetate promoted PYY secretion in STC-1 cells via Olfr78-protein kinase A signaling, whereas the effects were abolished by Olfr78 RNA interference. Colonic SCFAs production via oral administration of fructo-oligosaccharide significantly increased plasma PYY levels, whereas this effect was abolished in Olfr78-deficient and germ-free mice. These results suggested that the SCFA receptor Olfr78 is important for anti-obesity and anorexigenic effects of the gut microbiota and dietary fibers.


Anorexia/metabolism , Anorexia/microbiology , Fatty Acids, Volatile/pharmacology , Gastrointestinal Microbiome , Intestinal Mucosa/microbiology , Obesity/microbiology , Peptide YY/metabolism , Receptors, Odorant/metabolism , Animals , Anorexia/pathology , Cells, Cultured , Disease Models, Animal , Enteroendocrine Cells/metabolism , Intestinal Mucosa/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/metabolism , Obesity/pathology
14.
Nephrol Dial Transplant ; 35(9): 1501-1517, 2020 09 01.
Article En | MEDLINE | ID: mdl-32535631

BACKGROUND: Chronic kidney disease (CKD) leads to insulin resistance (IR) and sarcopenia, which are associated with a high mortality risk in CKD patients; however, their pathophysiologies remain unclear. Recently, alterations in gut microbiota have been reported to be associated with CKD. We aimed to determine whether uremic dysbiosis contributes to CKD-associated IR and sarcopenia. METHODS: CKD was induced in specific pathogen-free mice via an adenine-containing diet; control animals were fed a normal diet. Fecal microbiota transplantation (FMT) was performed by oral gavage in healthy germ-free mice using cecal bacterial samples obtained from either control mice (control-FMT) or CKD mice (CKD-FMT). Vehicle mice were gavaged with sterile phosphate-buffered saline. Two weeks after inoculation, mice phenotypes, including IR and sarcopenia, were evaluated. RESULTS: IR and sarcopenia were evident in CKD mice compared with control mice. These features were reproduced in CKD-FMT mice compared with control-FMT and vehicle mice with attenuated insulin-induced signal transduction and mitochondrial dysfunction in skeletal muscles. Intestinal tight junction protein expression and adipocyte sizes were lower in CKD-FMT mice than in control-FMT mice. Furthermore, CKD-FMT mice showed systemic microinflammation, increased concentrations of serum uremic solutes, fecal bacterial fermentation products and elevated lipid content in skeletal muscle. The differences in gut microbiota between CKD and control mice were mostly consistent between CKD-FMT and control-FMT mice. CONCLUSIONS: Uremic dysbiosis induces IR and sarcopenia, leaky gut and lipodystrophy.


Bacteria/isolation & purification , Dysbiosis/complications , Gastrointestinal Microbiome , Insulin Resistance , Renal Insufficiency, Chronic/physiopathology , Sarcopenia/etiology , Uremia/complications , Animals , Dysbiosis/microbiology , Fecal Microbiota Transplantation , Lipids/blood , Male , Mice , Mice, Inbred ICR , Sarcopenia/pathology , Sarcopenia/therapy
15.
Science ; 367(6481)2020 02 28.
Article En | MEDLINE | ID: mdl-32108090

Antibiotics and dietary habits can affect the gut microbial community, thus influencing disease susceptibility. Although the effect of microbiota on the postnatal environment has been well documented, much less is known regarding the impact of gut microbiota at the embryonic stage. Here we show that maternal microbiota shapes the metabolic system of offspring in mice. During pregnancy, short-chain fatty acids produced by the maternal microbiota dictate the differentiation of neural, intestinal, and pancreatic cells through embryonic GPR41 and GPR43. This developmental process helps maintain postnatal energy homeostasis, as evidenced by the fact that offspring from germ-free mothers are highly susceptible to metabolic syndrome, even when reared under conventional conditions. Thus, our findings elaborate on a link between the maternal gut environment and the developmental origin of metabolic syndrome.

16.
Proc Natl Acad Sci U S A ; 116(47): 23813-23821, 2019 11 19.
Article En | MEDLINE | ID: mdl-31685604

Ketone bodies, including ß-hydroxybutyrate and acetoacetate, are important alternative energy sources during energy shortage. ß-Hydroxybutyrate also acts as a signaling molecule via specific G protein-coupled receptors (GPCRs); however, the specific associated GPCRs and physiological functions of acetoacetate remain unknown. Here we identified acetoacetate as an endogenous agonist for short-chain fatty acid (SCFA) receptor GPR43 by ligand screening in a heterologous expression system. Under ketogenic conditions, such as starvation and low-carbohydrate diets, plasma acetoacetate levels increased markedly, whereas plasma and cecal SCFA levels decreased dramatically, along with an altered gut microbiota composition. In addition, Gpr43-deficient mice showed reduced weight loss and suppressed plasma lipoprotein lipase activity during fasting and eucaloric ketogenic diet feeding. Moreover, Gpr43-deficient mice exhibited minimal weight decrease after intermittent fasting. These observations provide insight into the role of ketone bodies in energy metabolism under shifts in nutrition and may contribute to the development of preventive medicine via diet and foods.


Diet, Ketogenic , Ketone Bodies/metabolism , Lipid Metabolism/physiology , Receptors, G-Protein-Coupled/physiology , Animals , Fasting , HEK293 Cells , Humans , Ligands , Lipoprotein Lipase/blood , Mice , Mice, Inbred C57BL , Receptors, G-Protein-Coupled/genetics , Signal Transduction
17.
Int J Mol Sci ; 20(18)2019 Sep 10.
Article En | MEDLINE | ID: mdl-31509948

As glucose-dependent insulinotropic polypeptide (GIP) possesses pro-adipogenic action, the suppression of the GIP hypersecretion seen in obesity might represent a novel therapeutic approach to the treatment of obesity. However, the mechanism of GIP hypersecretion remains largely unknown. In the present study, we investigated GIP secretion in two mouse models of obesity: High-fat diet-induced obese (DIO) mice and leptin-deficient Lepob/ob mice. In DIO mice, plasma GIP was increased along with an increase in GIP mRNA expression in the lower small intestine. Despite the robust alteration in the gut microbiome in DIO mice, co-administration of maltose and the α-glucosidase inhibitor (α-GI) miglitol induced the microbiome-mediated suppression of GIP secretion. The plasma GIP levels of Lepob/ob mice were also elevated and were suppressed by fat transplantation. The GIP mRNA expression in fat tissue was not increased in Lepob/ob mice, while the expression of an interleukin-1 receptor antagonist (IL-1Ra) was increased. Fat transplantation suppressed the expression of IL-1Ra. The plasma IL-1Ra levels were positively correlated with the plasma GIP levels. Accordingly, although circulating GIP levels are increased in both DIO and Lepob/ob mice, the underlying mechanisms differ, and the anti-obesity actions of α-GIs and leptin sensitizers may be mediated partly by the suppression of GIP secretion.


Diet, High-Fat/adverse effects , Gastric Inhibitory Polypeptide/metabolism , Leptin/deficiency , Obesity/metabolism , Animals , Gastric Inhibitory Polypeptide/blood , Gastric Inhibitory Polypeptide/genetics , Gene Expression , Interleukin 1 Receptor Antagonist Protein/blood , Interleukin 1 Receptor Antagonist Protein/genetics , Interleukin 1 Receptor Antagonist Protein/metabolism , Leptin/genetics , Male , Mice, Inbred C57BL , Mice, Obese , Obesity/etiology , Obesity/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Gastrointestinal Hormone/genetics , Receptors, Gastrointestinal Hormone/metabolism
18.
Nat Commun ; 10(1): 4007, 2019 09 05.
Article En | MEDLINE | ID: mdl-31488836

Gut microbiota mediates the effects of diet, thereby modifying host metabolism and the incidence of metabolic disorders. Increased consumption of omega-6 polyunsaturated fatty acid (PUFA) that is abundant in Western diet contributes to obesity and related diseases. Although gut-microbiota-related metabolic pathways of dietary PUFAs were recently elucidated, the effects on host physiological function remain unclear. Here, we demonstrate that gut microbiota confers host resistance to high-fat diet (HFD)-induced obesity by modulating dietary PUFAs metabolism. Supplementation of 10-hydroxy-cis-12-octadecenoic acid (HYA), an initial linoleic acid-related gut-microbial metabolite, attenuates HFD-induced obesity in mice without eliciting arachidonic acid-mediated adipose inflammation and by improving metabolic condition via free fatty acid receptors. Moreover, Lactobacillus-colonized mice show similar effects with elevated HYA levels. Our findings illustrate the interplay between gut microbiota and host energy metabolism via the metabolites of dietary omega-6-FAs thereby shedding light on the prevention and treatment of metabolic disorders by targeting gut microbial metabolites.


Diet, High-Fat/adverse effects , Dietary Fats, Unsaturated/therapeutic use , Fatty Acids, Unsaturated/pharmacology , Gastrointestinal Microbiome/drug effects , Obesity/metabolism , Adipose Tissue/pathology , Animals , Cell Line , Diet, Western , Dietary Supplements , Energy Metabolism , Fatty Acids, Omega-6/metabolism , Fatty Acids, Omega-6/therapeutic use , Fatty Acids, Unsaturated/metabolism , Gastrointestinal Microbiome/physiology , Humans , Inflammation/metabolism , Lactobacillus/metabolism , Linoleic Acid/metabolism , Metabolic Diseases/diet therapy , Metabolic Diseases/metabolism , Metabolic Diseases/prevention & control , Mice , Mice, Inbred C57BL , Models, Animal , Oleic Acids/metabolism
19.
Nutrients ; 11(5)2019 May 09.
Article En | MEDLINE | ID: mdl-31075850

4-Hydroxy-3-methoxycinnamic acid (HMCA), a hydroxycinnamic acid derivative, is abundant in fruits and vegetables, including oranges, carrots, rice bran, and coffee beans. Several beneficial effects of HMCA have been reported, including improvement of metabolic abnormalities in animal models and human studies. However, its mitigating effects on high-fat diet (HFD)-induced obesity, and the mechanism underlying these effects, remain to be elucidated. In this study, we demonstrated that dietary HMCA was efficacious against HFD-induced weight gain and hepatic steatosis, and that it improved insulin sensitivity. These metabolic benefits of HMCA were ascribable to 3-(4-hydroxy-3-methoxyphenyl)propionic acid (HMPA) produced by gut microbiota. Moreover, conversion of HMCA into HMPA was attributable to a wide variety of microbes belonging to the phylum Bacteroidetes. We further showed that HMPA modulated gut microbes associated with host metabolic homeostasis by increasing the abundance of organisms belonging to the phylum Bacteroidetes and reducing the abundance of the phylum Firmicutes. Collectively, these results suggest that HMPA derived from HMCA is metabolically beneficial, and regulates hepatic lipid metabolism, insulin sensitivity, and the gut microbial community. Our results provide insights for the development of functional foods and preventive medicines, based on the microbiota of the intestinal environment, for the prevention of metabolic disorders.


Coumaric Acids/pharmacology , Diet , Gastrointestinal Microbiome/drug effects , Lipid Metabolism/drug effects , Liver/drug effects , Obesity/metabolism , Propionates/pharmacology , Animals , Bacteroidetes/drug effects , Bacteroidetes/growth & development , Bacteroidetes/metabolism , Citrus sinensis/chemistry , Coffea/chemistry , Coumaric Acids/metabolism , Daucus carota/chemistry , Diet, High-Fat/adverse effects , Fatty Liver/etiology , Fatty Liver/prevention & control , Firmicutes/growth & development , Firmicutes/metabolism , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/microbiology , Insulin Resistance , Liver/metabolism , Liver/pathology , Male , Metabolic Diseases/etiology , Metabolic Diseases/metabolism , Metabolic Diseases/pathology , Metabolic Diseases/prevention & control , Mice, Inbred C57BL , Mice, Obese , Obesity/complications , Obesity/etiology , Oryza/chemistry , Plants, Edible/chemistry , Propionates/metabolism , Weight Gain/drug effects
20.
J Endocrinol ; 239(3): 267-276, 2018 12 01.
Article En | MEDLINE | ID: mdl-30400014

Mechanisms of carbohydrate-induced secretion of the two incretins namely glucagon-like peptide 1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) are considered to be mostly similar. However, we found that mice exhibit opposite secretory responses in response to co-administration of maltose plus an α-glucosidase inhibitor miglitol (maltose/miglitol), stimulatory for GLP-1, as reported previously, but inhibitory for GIP. Gut microbiota was shown to be involved in maltose/miglitol-induced GIP suppression, as the suppression was attenuated in antibiotics (Abs)-treated mice and abolished in germ-free mice. In addition, maltose/miglitol administration increased plasma levels of short-chain fatty acids (SCFAs), carbohydrate-derived metabolites, in the portal vein. GIP suppression by maltose/miglitol was not observed in mice lacking a SCFA receptor Ffar3, but it was normally seen in Ffar2-deficient mice. Similar to maltose/miglitol administration, co-administration of glucose plus a sodium glucose transporter inhibitor phloridzin (glucose/phloridzin) induced GIP suppression, which was again cancelled by Abs treatment. In conclusion, oral administration of carbohydrates with α-glucosidase inhibitors suppresses GIP secretion through a microbiota/SCFA/FFAR3 pathway.


Gastric Inhibitory Polypeptide/metabolism , Glucagon-Like Peptide 1/metabolism , 1-Deoxynojirimycin/analogs & derivatives , Animals , Carbohydrate Metabolism , Fatty Acids, Volatile/metabolism , Gastrointestinal Microbiome/physiology , Glycoside Hydrolase Inhibitors , Incretins/metabolism , KATP Channels/metabolism , Maltose , Mice , Receptors, G-Protein-Coupled/metabolism
...