Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 103
1.
Gut Microbes ; 16(1): 2359729, 2024.
Article En | MEDLINE | ID: mdl-38816999

Early life environment influences mammalian brain development, a growing area of research within the Developmental Origins of Health and Disease framework, necessitating a deeper understanding of early life factors on children's brain development. This study introduces a mouse model, LAO1 knockout mice, to investigate the relationship between breast milk, the gut microbiome, and brain development. The results reveal that breast milk's reactive oxygen species (ROS) are vital in shaping the neonatal gut microbiota. Decreased hydrogen peroxide (H2O2) levels in milk disrupt the gut microbiome and lead to abnormal metabolite production, including D-glucaric acid. This metabolite inhibits hippocampal myelin formation during infancy, potentially contributing to behavioral abnormalities observed in adulthood. These findings suggest that H2O2 in breast milk is crucial for normal gut microbiota formation and brain development, with implications for understanding and potentially treating neurodevelopmental disorders in humans.


Animals, Newborn , Gastrointestinal Microbiome , Hydrogen Peroxide , Mice, Knockout , Milk, Human , Myelin Sheath , Animals , Myelin Sheath/metabolism , Mice , Animals, Newborn/growth & development , Milk, Human/chemistry , Milk, Human/metabolism , Humans , Female , Hydrogen Peroxide/metabolism , Brain/metabolism , Brain/growth & development , Reactive Oxygen Species/metabolism , Hippocampus/metabolism , Hippocampus/growth & development , Male , Mice, Inbred C57BL
2.
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
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.
Brain Sci ; 14(3)2024 Feb 21.
Article En | MEDLINE | ID: mdl-38539586

In recent years, neurorehabilitation has been actively used to treat motor paralysis after stroke. However, the impacts of rehabilitation on neural networks in the brain remain largely unknown. Therefore, we investigated changes in structural neural networks after rehabilitation therapy in patients who received a combination of low-frequency repetitive transcranial magnetic stimulation (LF-rTMS) and intensive occupational therapy (intensive-OT) as neurorehabilitation. Fugl-Meyer assessment (FMA) for upper extremity (FMA-UE) and Action Research Arm Test (ARAT), both of which reflected upper limb motor function, were conducted before and after rehabilitation therapy. At the same time, diffusion tensor imaging (DTI) and three-dimensional T1-weighted imaging (3D T1WI) were performed. After analyzing the structural connectome based on DTI data, measures related to connectivity in neural networks were calculated using graph theory. Rehabilitation therapy prompted a significant increase in connectivity with the isthmus of the cingulate gyrus in the ipsilesional hemisphere (p < 0.05) in patients with left-sided paralysis, as well as a significant decrease in connectivity with the ipsilesional postcentral gyrus (p < 0.05). These results indicate that LF-rTMS combined with intensive-OT may facilitate motor function recovery by enhancing the functional roles of networks in motor-related areas of the ipsilesional cerebral hemisphere.

5.
Sci Rep ; 14(1): 1056, 2024 01 11.
Article En | MEDLINE | ID: mdl-38212379

The gut microbiota has emerged as an important factor that potentially influences various physiological functions and pathophysiological processes such as obesity and type 2 diabetes mellitus. Accumulating evidence from human and animal studies suggests that gut microbial metabolites play a critical role as integral molecules in host-microbe interactions. Notably, several dietary environment-dependent fatty acid metabolites have been recognized as potent modulators of host metabolic homeostasis. More recently, nicotine, the primary active molecule in tobacco, has been shown to potentially affect host metabolism through alterations in the gut microbiota and its metabolites. However, the mechanisms underlying the interplay between host nutritional status, diet-derived microbial metabolites, and metabolic homeostasis during nicotine exposure remain unclear. Our findings revealed that nicotine administration had potential effects on weight regulation and metabolic phenotype, independent of reduced caloric intake. Moreover, nicotine-induced body weight suppression is associated with specific changes in gut microbial composition, including Lactobacillus spp., and KetoB, a nicotine-sensitive gut microbiota metabolite, which could be linked to changes in host body weight, suggesting its potential role in modulating host metabolism. Our findings highlight the remarkable impact of the interplay between nutritional control and the gut environment on host metabolism during smoking and smoking cessation.


Diabetes Mellitus, Type 2 , Gastrointestinal Microbiome , Animals , Humans , Gastrointestinal Microbiome/physiology , Nicotine/pharmacology , Diet , Obesity/etiology
6.
Sci Rep ; 13(1): 21246, 2023 12 01.
Article En | MEDLINE | ID: mdl-38040866

3-(4-hydroxy-3-methoxyphenyl) propionic acid (HMPA) is a metabolite produced by the gut microbiota through the conversion of 4-hydroxy-3-methoxycinnamic acid (HMCA), which is a widely distributed hydroxycinnamic acid-derived metabolite found abundantly in plants. Several beneficial effects of HMPA have been suggested, such as antidiabetic properties, anticancer activities, and cognitive function improvement, in animal models and human studies. However, the intricate molecular mechanisms underlying the bioaccessibility and bioavailability profile following HMPA intake and the substantial modulation of metabolic homeostasis by HMPA require further elucidation. In this study, we effectively identified and characterized HMPA-specific GPR41 receptor, with greater affinity than HMCA. The activation of this receptor plays a crucial role in the anti-obesity effects and improvement of hepatic steatosis by stimulating the lipid catabolism pathway. For the improvement of metabolic disorders, our results provide insights into the development of functional foods, including HMPA, and preventive pharmaceuticals targeting GPR41.


Hempa , Lipid Metabolism , Animals , Humans , Hempa/metabolism , Liver/metabolism , Propionates/pharmacology , Propionates/metabolism
7.
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
8.
Glia ; 71(11): 2609-2622, 2023 11.
Article En | MEDLINE | ID: mdl-37470163

Resident microglia are important to maintain homeostasis in the central nervous system, which includes the retina. The retinal microglia become activated in numerous pathological conditions, but the molecular signatures of these changes are poorly understood. Here, using an approach based on FACS and RNA-seq, we show that microglial gene expression patterns gradually change during RGC degeneration induced by optic nerve injury. Most importantly, we found that the microglial cells strongly expressed Tnf and Il1α, both of which are known to induce neurotoxic reactive astrocytes, and were characterized by Gpr84high -expressing cells in a particular subpopulation. Moreover, ripasudil, a Rho kinase inhibitor, significantly blunted Gpr84 expression and cytokine induction in vitro and in vivo. Finally, GPR84-deficient mice prevented RGC loss in optic nerve-injured retina. These results reveal that Rho kinase-mediated GPR84 alteration strongly contribute to microglial activation and promote neurotoxicity, suggesting that Rho-ROCK and GPR84 signaling may be potential therapeutic targets to prevent the neurotoxic microglial phenotype induced by optic nerve damage, such as occurs in traumatic optic neuropathy and glaucoma.


Optic Nerve Injuries , Mice , Animals , Microglia/metabolism , Retinal Ganglion Cells , rho-Associated Kinases/metabolism , Neuroglia/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism
9.
Nat Commun ; 14(1): 3863, 2023 06 30.
Article En | MEDLINE | ID: mdl-37391427

Fever is a common symptom of influenza and coronavirus disease 2019 (COVID-19), yet its physiological role in host resistance to viral infection remains less clear. Here, we demonstrate that exposure of mice to the high ambient temperature of 36 °C increases host resistance to viral pathogens including influenza virus and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). High heat-exposed mice increase basal body temperature over 38 °C to enable more bile acids production in a gut microbiota-dependent manner. The gut microbiota-derived deoxycholic acid (DCA) and its plasma membrane-bound receptor Takeda G-protein-coupled receptor 5 (TGR5) signaling increase host resistance to influenza virus infection by suppressing virus replication and neutrophil-dependent tissue damage. Furthermore, the DCA and its nuclear farnesoid X receptor (FXR) agonist protect Syrian hamsters from lethal SARS-CoV-2 infection. Moreover, we demonstrate that certain bile acids are reduced in the plasma of COVID-19 patients who develop moderate I/II disease compared with the minor severity of illness group. These findings implicate a mechanism by which virus-induced high fever increases host resistance to influenza virus and SARS-CoV-2 in a gut microbiota-dependent manner.


COVID-19 , Gastrointestinal Microbiome , Influenza A virus , Influenza, Human , Cricetinae , Animals , Mice , Humans , SARS-CoV-2 , Body Temperature , Fever , Bile Acids and Salts , Mesocricetus
10.
Gut Microbes ; 15(1): 2206507, 2023.
Article En | MEDLINE | ID: mdl-37131293

Evidence has accumulated that gut microbiota and its metabolites, in particular the short-chain fatty acid propionate, are significant contributors to the pathogenesis of a variety of diseases. However, little is known regarding its impact on pediatric bronchial asthma, one of the most common allergic diseases in childhood. This study aimed to elucidate whether, and if so how, intestinal propionate during lactation is involved in the development of bronchial asthma. We found that propionate intake through breast milk during the lactation period resulted in a significant reduction of airway inflammation in the offspring in a murine house dust mite-induced asthma model. Moreover, GPR41 was the propionate receptor involved in suppressing this asthmatic phenotype, likely through the upregulation of Toll-like receptors. In translational studies in a human birth cohort, we found that fecal propionate was decreased one month after birth in the group that later developed bronchial asthma. These findings indicate an important role for propionate in regulating immune function to prevent the pathogenesis of bronchial asthma in childhood.


Asthma , Gastrointestinal Microbiome , Female , Humans , Infant , Child , Animals , Mice , Propionates , Asthma/prevention & control , Fatty Acids, Volatile/metabolism , Intestines , Disease Susceptibility
11.
Nat Chem Biol ; 19(6): 778-789, 2023 06.
Article En | MEDLINE | ID: mdl-36864192

Mucinolytic bacteria modulate host-microbiota symbiosis and dysbiosis through their ability to degrade mucin O-glycans. However, how and to what extent bacterial enzymes are involved in the breakdown process remains poorly understood. Here we focus on a glycoside hydrolase family 20 sulfoglycosidase (BbhII) from Bifidobacterium bifidum, which releases N-acetylglucosamine-6-sulfate from sulfated mucins. Glycomic analysis showed that, in addition to sulfatases, sulfoglycosidases are involved in mucin O-glycan breakdown in vivo and that the released N-acetylglucosamine-6-sulfate potentially affects gut microbial metabolism, both of which were also supported by a metagenomic data mining analysis. Enzymatic and structural analysis of BbhII reveals the architecture underlying its specificity and the presence of a GlcNAc-6S-specific carbohydrate-binding module (CBM) 32 with a distinct sugar recognition mode that B. bifidum takes advantage of to degrade mucin O-glycans. Comparative analysis of the genomes of prominent mucinolytic bacteria also highlights a CBM-dependent O-glycan breakdown strategy used by B. bifidum.


Ecosystem , Mucins , Mucins/metabolism , Polysaccharides/metabolism , Bacteria/metabolism
12.
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
13.
Mol Metab ; 67: 101649, 2023 01.
Article En | MEDLINE | ID: mdl-36462626

OBJECTIVE: The gastrointestinal tract affects physiological activities and behavior by secreting hormones and generating signals through the activation of nutrient sensors. GPR119, a lipid sensor, is indirectly involved in the secretion of incretins, such as glucagon-like peptide-1 and glucose-dependent insulinotropic peptide, by enteroendocrine cells, while it directly stimulates insulin secretion by pancreatic beta cells. Since GPR119 has the potential to modulate metabolic homeostasis in obesity and diabetes, it has attracted interest as a therapeutic target. However, previous studies have shown that the deletion of Gpr119 in mice does not affect glucose homeostasis and appetite in either basal or high-fat diet-fed conditions. Therefore, the present study aimed to explore the role of GPR119 signaling system in energy metabolism and feeding behavior in mice. METHODS: Gpr119 knockout (KO) mice were generated using CRISPR-Cas9 gene-editing technology, and their feeding behavior and energy metabolism were evaluated and compared with those of wild type (WT) mice. RESULTS: Upon inducing metabolic stress via food deprivation, Gpr119 KO mice exhibited lower blood glucose levels and a higher body weight reduction compared to WT mice. Although food intake in WT and KO mice were similar under free-feeding conditions, Gpr119 KO mice exhibited increased food intake when they were refed after 24 h of food deprivation. Further, food-deprived Gpr119 KO mice presented shorter post-meal intervals and lower satiety for second and later meals during refeeding, resulting in increased food intake. Associated with this meal pattern, levels of oleoylethanolamide (OEA), an endogenous agonist of GPR119, in the luminal contents of the distal gastrointestinal tract were elevated within 2 h after refeeding. The large-intestinal infusion of OEA prolonged post-meal intervals and increased satiety in the first meal, but not the second meal. On the other hand, infusion of oleic acid increased cecal OEA levels at 2 h from the beginning of infusion, while prolonging post-meal intervals and increasing satiety on the meals that occurred approximately 2 h after the infusion. Cecal OEA levels were low in antibiotic-treated mice, suggesting that the gut microbiota partially synthesizes OEA from oleic acid. CONCLUSIONS: Collectively, our results indicate that the activation of gastrointestinal GPR119 by microbiota-produced OEA derived from oleic acid is associated with satiety control and energy homeostasis under energy shortage conditions.


Microbiota , Oleic Acid , Mice , Animals , Feeding Behavior , Mice, Knockout , Energy Metabolism
14.
JCI Insight ; 8(2)2023 Jan 24.
Article En | MEDLINE | ID: mdl-36480287

Medium-chain triglycerides (MCTs), which consist of medium-chain fatty acids (MCFAs), are unique forms of dietary fat with various health benefits. G protein-coupled 84 (GPR84) acts as a receptor for MCFAs (especially C10:0 and C12:0); however, GPR84 is still considered an orphan receptor, and the nutritional signaling of endogenous and dietary MCFAs via GPR84 remains unclear. Here, we showed that endogenous MCFA-mediated GPR84 signaling protected hepatic functions from diet-induced lipotoxicity. Under high-fat diet (HFD) conditions, GPR84-deficient mice exhibited nonalcoholic steatohepatitis (NASH) and the progression of hepatic fibrosis but not steatosis. With markedly increased hepatic MCFA levels under HFD, GPR84 suppressed lipotoxicity-induced macrophage overactivation. Thus, GPR84 is an immunomodulating receptor that suppresses excessive dietary fat intake-induced toxicity by sensing increases in MCFAs. Additionally, administering MCTs, MCFAs (C10:0 or C12:0, but not C8:0), or GPR84 agonists effectively improved NASH in mouse models. Therefore, exogenous GPR84 stimulation is a potential strategy for treating NASH.


Non-alcoholic Fatty Liver Disease , Receptors, G-Protein-Coupled , Mice , Animals , Receptors, G-Protein-Coupled/agonists , Fatty Acids , Dietary Fats/pharmacology , Triglycerides , Liver Cirrhosis
15.
Mol Nutr Food Res ; 66(22): e2200063, 2022 11.
Article En | MEDLINE | ID: mdl-36181445

SCOPE: The purpose of this study is to compare the impact of four low-viscosity soluble dietary fibers (DFs) on the intestinal microenvironment, in terms of microbiota composition, short-chain fatty acid (SCFA) production, proportion of colonic peripherally induced regulatory T cells (pTregs), and experimental colitis in mice. METHODS AND RESULTS: Mice are administered 5% w/v low-viscosity soluble DFs in drinking water for 2 weeks. The gut microbiota composition is determined using 16S rRNA sequencing. Luminal SCFAs are quantified by gas chromatography, and colonic pTregs are analyzed using flow cytometry. All low-viscosity soluble DFs promote the growth of beneficial bacteria such as Akkermansia muciniphila and Bacteroides acidifaciens, while eliminating pathogenic bacteria such as Clostridium perfringens. Moreover, two low-viscosity soluble DFs significantly increase the abundance of commensal bacteria and promote the accumulation of propionate and butyrate, leading to marked induction of colonic pTregs. Consistently, these two fibers, in particular α-cyclodextrin, show remarkable anti-inflammatory properties in a colitis mouse model. CONCLUSION: Mice administered any low-viscosity soluble DF show comparable gut microbiota compositions, but differ in terms of bacterial abundance, SCFA concentration, pTreg population, and colitis development. This exploratory study suggests that administration of α-cyclodextrin may be a possible strategy for the prevention of colitis.


Colitis , alpha-Cyclodextrins , Mice , Animals , RNA, Ribosomal, 16S/genetics , Viscosity , Colitis/microbiology , Verrucomicrobia , Fatty Acids, Volatile/analysis , Dietary Fiber/pharmacology , Inflammation/prevention & control , Mice, Inbred C57BL
16.
Food Funct ; 13(21): 10970-10980, 2022 Oct 31.
Article En | MEDLINE | ID: mdl-36254783

Barley consumption is expected to increase insulin sensitivity by increasing the level of short-chain fatty acids (SCFAs) and promoting the secretion of GLP-1. However, the involvement of GPR43, a receptor for SCFAs, has not been investigated. Therefore, we evaluated whether the inhibitory effect of ß-glucan-rich barley intake on blood glucose rise is mediated by GPR43 signalling via an increase of SCFAs. C57BL/6J mice and GPR43-knockout mice were fed high-fat diets with either cellulose (HC) or ß-glucan-rich barley flour (HB) for 12 weeks. The level of SCFAs in cecum contents was measured and the concentration of GLP-1 in the portal vein was determined. The supernatant of the cecum contents of C57BL/6J mice was added to GLUTag cells, and then the changes to GLP-1 and intracellular Ca2+ concentrations determined. The same parameters were measured using cells in which GPR43 was knocked down by siRNA. C57BL/6J mice fed HB diets showed a suppressed glucose rise compared to those on the HC diet. Cecum SCFAs and GLP-1 concentration in the portal vein were also increased by the HB diet. When an aqueous solution from the cecum content of mice fed a HB diet was added to GLUTag cells, GLP-1 secretion and intracellular Ca2+ concentration were increased. These phenomena were not observed in cells with knockdown of GPR43. In GPR43 knockout mice an increase of GLP-1 in the portal vein and suppression of blood glucose elevation was attenuated, despite increased SCFAs brought on by the HB diet. In conclusion, GPR43 activation in the intestinal tract via increased SCFAs is required for the glucose intolerance-improving effect of barley consumption.


Glucose Intolerance , Hordeum , beta-Glucans , Mice , Male , Animals , Hordeum/metabolism , Mice, Obese , Blood Glucose , Flour , Fermentation , Mice, Inbred C57BL , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Fatty Acids, Volatile , Glucagon-Like Peptide 1 , beta-Glucans/pharmacology , Mice, Knockout , Obesity
17.
Pharmacol Ther ; 239: 108273, 2022 11.
Article En | MEDLINE | ID: mdl-36057320

Dysbiosis is associated with various diseases. The composition and diversity of gut microbiota affect host physiology through the production of bioactive metabolites. Short-chain fatty acids are the main metabolites produced by microbial fermentation of dietary fiber. They play a crucial role in maintaining metabolic, nervous, and immune system. Short-chain fatty acids not only serve as an energy source for the host but also act as for G-protein-coupled receptor signaling molecules and histone deacetylase inhibitors. In particular, the discovery and deorphanization of free fatty acid receptors 2 and 3 (GPR43/41) have shed light on the molecular mechanisms underlying the regulation of physiological processes by short-chain fatty acids. The short-chain fatty acid receptors sense the nutrient status and transduce signals to maintain cellular homeostasis. Dysbiosis affects short-chain fatty acid production and impairs the signaling, leading to cellular dysfunction. We review the current understanding of short-chain fatty acid-mediated regulation of physiological processes and discuss the molecular pharmacology of short-chain fatty acid and the receptor. We also discuss recent advances in the use of prebiotics and probiotics in the treatment of disease.


Gastrointestinal Microbiome , Humans , Dysbiosis/complications , Fatty Acids, Volatile/metabolism , Energy Metabolism , Dietary Fiber
18.
Sci Rep ; 12(1): 13819, 2022 08 15.
Article En | MEDLINE | ID: mdl-35970869

The gut environment, including the microbiota and its metabolites and short-chain fatty acids (SCFA), is essential for health maintenance. It is considered that functional recovery treatment for masticatory dysphagia affects the composition of the gut microbiota, indicating that habitual mastication, depending on the hardness of the food, may affect the gut microbiota and environment. However, the impact of chronic powdered diet feeding on the colonic condition and motility remains unclear. Here, we evaluated various colonic features in mice fed with powdered diets for a long-term and a mouse model with masticatory behavior. We observed a decreased abundance of the SCFA-producing bacterial genera in the ceca of the powdered diet-fed mice. Based on the importance of SCFAs in gut immune homeostasis and motility, interestingly, powdered diet feeding also resulted in constipation-like symptoms due to mild colitis, which were ameliorated by the administration of a neutrophil-depleting agent and neutrophil elastase inhibitors. Lastly, the suppressed colonic motility in the powdered diet-fed mice was significantly improved by loading masticatory activity for 2 h. Thus, feeding habits with appropriate masticatory activity and stimulation may play a key role in providing a favorable gut environment based on interactions between the gut microbiota and host immune system.


Colitis , Gastrointestinal Microbiome , Microbiota , Animals , Fatty Acids, Volatile/metabolism , Mastication , Mice
19.
Cell Rep ; 40(3): 111087, 2022 07 19.
Article En | MEDLINE | ID: mdl-35858544

Microbiota-accessible carbohydrates (MACs) exert health-promoting effects, but how each MAC impacts gut microbiota and regulates host physiology remains unclear. Here, we show that l-arabinose and sucrose cooperatively act on gut microbiota and exert anti-obesogenic effects. Specifically, l-arabinose, a monosaccharide that is poorly absorbed in the gut and inhibits intestinal sucrase, suppresses diet-induced obesity in mice in the presence of sucrose. Additionally, the suppressive effect of l-arabinose on adiposity is abrogated in mice lacking the short-chain fatty acid (SCFA) receptors GPR43 and GPR41. Mechanistically, l-arabinose increases the relative abundance of acetate and propionate producers (e.g., Bacteroides), while sucrose enhances SCFA production. Furthermore, l-arabinose and sucrose activate the glycolytic and pentose phosphate pathways of Bacteroides, respectively, indicating that they synergistically promote acetate production through distinct pathways. These findings suggest that each MAC has a unique property and thus may serve as a precision gut-microbiota modulator to promote host homeostasis.


Gastrointestinal Microbiome , Microbiota , Animals , Arabinose/pharmacology , Bacteroides/metabolism , Carbohydrates , Fatty Acids, Volatile/metabolism , Mice , Obesity/metabolism , Sucrose
20.
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
...