Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Cell Host Microbe ; 32(5): 630-632, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38723600

ABSTRACT

The gut microbiota has the capacity to metabolize food-derived molecules. In this issue of Cell Host & Microbe, Li et al. explore how some bacterial species of the gut microbiota can deplete amino acids in the gut lumen, modulating the amino acid landscape and energy metabolism of the host.


Subject(s)
Amino Acids , Energy Metabolism , Gastrointestinal Microbiome , Gastrointestinal Microbiome/physiology , Amino Acids/metabolism , Humans , Bacteria/metabolism , Bacteria/genetics , Animals , Host Microbial Interactions , Gastrointestinal Tract/microbiology
2.
Science ; 379(6634): 826-833, 2023 02 24.
Article in English | MEDLINE | ID: mdl-36821686

ABSTRACT

The intestinal microbiota is known to influence postnatal growth. We previously found that a strain of Lactiplantibacillus plantarum (strain LpWJL) buffers the adverse effects of chronic undernutrition on the growth of juvenile germ-free mice. Here, we report that LpWJL sustains the postnatal growth of malnourished conventional animals and supports both insulin-like growth factor-1 (IGF-1) and insulin production and activity. We have identified cell walls isolated from LpWJL, as well as muramyl dipeptide and mifamurtide, as sufficient cues to stimulate animal growth despite undernutrition. Further, we found that NOD2 is necessary in intestinal epithelial cells for LpWJL-mediated IGF-1 production and for postnatal growth promotion in malnourished conventional animals. These findings indicate that, coupled with renutrition, bacteria cell walls or purified NOD2 ligands have the potential to alleviate stunting.


Subject(s)
Gastrointestinal Microbiome , Growth , Intestines , Lactobacillaceae , Malnutrition , Nod2 Signaling Adaptor Protein , Animals , Mice , Cell Wall/chemistry , Epithelial Cells/microbiology , Epithelial Cells/physiology , Gastrointestinal Microbiome/physiology , Germ-Free Life , Growth Disorders/physiopathology , Growth Disorders/therapy , Insulin/metabolism , Insulin-Like Growth Factor I/metabolism , Intestinal Mucosa/microbiology , Intestinal Mucosa/physiology , Intestines/microbiology , Intestines/physiology , Lactobacillaceae/physiology , Malnutrition/physiopathology , Malnutrition/therapy , Nod2 Signaling Adaptor Protein/metabolism , Growth/drug effects , Growth/physiology , Acetylmuramyl-Alanyl-Isoglutamine/pharmacology , Acetylmuramyl-Alanyl-Isoglutamine/therapeutic use
3.
Nat Commun ; 12(1): 6686, 2021 11 18.
Article in English | MEDLINE | ID: mdl-34795236

ABSTRACT

Mus musculus is the classic mammalian model for biomedical research. Despite global efforts to standardize breeding and experimental procedures, the undefined composition and interindividual diversity of the microbiota of laboratory mice remains a limitation. In an attempt to standardize the gut microbiome in preclinical mouse studies, here we report the development of a simplified mouse microbiota composed of 15 strains from 7 of the 20 most prevalent bacterial families representative of the fecal microbiota of C57BL/6J Specific (and Opportunistic) Pathogen-Free (SPF/SOPF) animals and the derivation of a standardized gnotobiotic mouse model called GM15. GM15 recapitulates extensively the functionalities found in the C57BL/6J SOPF microbiota metagenome, and GM15 animals are phenotypically similar to SOPF or SPF animals in two different facilities. They are also less sensitive to the deleterious effects of post-weaning malnutrition. In this work, we show that the GM15 model provides increased reproducibility and robustness of preclinical studies by limiting the confounding effect of fluctuation in microbiota composition, and offers opportunities for research focused on how the microbiota shapes host physiology in health and disease.


Subject(s)
Feces/microbiology , Gastrointestinal Microbiome/physiology , Germ-Free Life , Specific Pathogen-Free Organisms , Whole Genome Sequencing/methods , Animals , Bacteria/classification , Bacteria/genetics , Body Weight/genetics , Body Weight/physiology , Female , Gastrointestinal Microbiome/genetics , Male , Metagenomics/methods , Mice, Inbred C57BL , Phenotype , Species Specificity
4.
J Mol Endocrinol ; 66(3): R67-R73, 2021 03.
Article in English | MEDLINE | ID: mdl-33410764

ABSTRACT

The worrying number of children suffering from undernutrition and consequent stunting worldwide makes the understanding of the relationship between nutritional status and postnatal growth crucial. Moreover, it is now well established that undernourished children harbor an altered microbiota, correlating with impaired growth. In this review, we describe how murine models have been used to explore the functional relationships between endocrine regulation of growth, nutrition and gut microbiota. In numerous Mammalian species, postnatal growth is mainly regulated by the conserved GH/IGF1 somatotropic axis that acts through endocrine and paracrine pathways, notably enabling longitudinal bone growth. Recent studies have demonstrated that the microbiota effects on growth could involve a modulation of GH and IGF1 circulating levels. Besides, the GH/IGF1 somatotropic axis may regulate the gut microbiota composition and diversity. Studying the bidirectional relationship between growth hormones and the gut microbiome could therefore help developing microbiota-targeting therapies in order to reduce the long-term consequences of stunting.


Subject(s)
Endocrine System/microbiology , Growth and Development , Nutritional Status , Animals , Gastrointestinal Microbiome , Growth Hormone/metabolism , Humans , Insulin-Like Growth Factor I/metabolism
5.
Trends Microbiol ; 29(8): 686-699, 2021 08.
Article in English | MEDLINE | ID: mdl-33309188

ABSTRACT

The gastrointestinal tract harbors an intrinsic neuronal network, the enteric nervous system (ENS). The ENS controls motility, fluid homeostasis, and blood flow, but also interacts with other components of the intestine such as epithelial and immune cells. Recent studies indicate that gut microbiota diversification, which occurs alongside postnatal ENS maturation, could be critical for the development and function of the ENS. Here we discuss the possibility that this functional relationship starts in utero, whereby the maternal microbiota would prime the developing ENS and shape its physiology. We review ENS/microbiota interactions and their modulation in physiological and pathophysiological contexts. While microbial modulation of the ENS physiology is now well established, further studies are required to understand the contribution of the gut microbiota to the development and pathology of the ENS and to reveal the precise mechanisms underlying microbiota-to-ENS communications.


Subject(s)
Enteric Nervous System/physiology , Gastrointestinal Microbiome/genetics , Gene Expression Regulation, Bacterial , Homeostasis , Enteric Nervous System/immunology , Enteric Nervous System/microbiology , Gastrointestinal Microbiome/physiology , Humans , Intestines/microbiology , Neurons/physiology
6.
Nutr Metab Insights ; 13: 1178638820980490, 2020.
Article in English | MEDLINE | ID: mdl-33402830

ABSTRACT

The gut microbiota is now recognized as a major contributor to the host's nutrition, metabolism, immunity, and neurological functions. Imbalanced microbiota (ie, dysbiosis) is linked to undernutrition-induced stunting, inflammatory and metabolic diseases, and cancers. Skeletal muscle also takes part in the interorgan crosstalk regulating substrate metabolism, immunity, and health. Here, we review the reciprocal influence of gut microbiota and skeletal muscle in relation to juvenile growth, performance, aging, and chronic diseases. Several routes involving the vascular system and organs such as the liver and adipose tissue connect the gut microbiota and skeletal muscle, with effects on fitness and health. Therapeutic perspectives arise from the health benefits observed with changes in gut microbiota and muscle activity, further encouraging multimodal therapeutic strategies.

7.
Proc Natl Acad Sci U S A ; 115(25): 6458-6463, 2018 06 19.
Article in English | MEDLINE | ID: mdl-29866843

ABSTRACT

The enteric nervous system (ENS) is crucial for essential gastrointestinal physiologic functions such as motility, fluid secretion, and blood flow. The gut is colonized by trillions of bacteria that regulate host production of several signaling molecules including serotonin (5-HT) and other hormones and neurotransmitters. Approximately 90% of 5-HT originates from the intestine, and activation of the 5-HT4 receptor in the ENS has been linked to adult neurogenesis and neuroprotection. Here, we tested the hypothesis that the gut microbiota could induce maturation of the adult ENS through release of 5-HT and activation of 5-HT4 receptors. Colonization of germ-free mice with a microbiota from conventionally raised mice modified the neuroanatomy of the ENS and increased intestinal transit rates, which was associated with neuronal and mucosal 5-HT production and the proliferation of enteric neuronal progenitors in the adult intestine. Pharmacological modulation of the 5-HT4 receptor, as well as depletion of endogenous 5-HT, identified a mechanistic link between the gut microbiota and maturation of the adult ENS through the release of 5-HT and activation of the 5-HT4 receptor. Taken together, these findings show that the microbiota modulates the anatomy of the adult ENS in a 5-HT-dependent fashion with concomitant changes in intestinal transit.


Subject(s)
Enteric Nervous System/microbiology , Enteric Nervous System/physiology , Gastrointestinal Microbiome/physiology , Intestine, Small/microbiology , Serotonin/metabolism , Animals , Enteric Nervous System/metabolism , Female , Gastrointestinal Motility/physiology , Intestine, Small/metabolism , Mice , Mice, Inbred C57BL , Microbiota/physiology , Neurogenesis/physiology , Neurons/metabolism , Neurons/microbiology , Receptors, Serotonin, 5-HT4/metabolism
8.
J Endocrinol ; 236(2): R105-R108, 2018 02.
Article in English | MEDLINE | ID: mdl-29321189

ABSTRACT

In the context of the obesity epidemic, dietary fibers that are found essentially in fruit and vegetables attract more and more attention, since they exert numerous metabolic benefits resulting in the moderation of body weight. Short-chain fatty acids, such as propionate and butyrate, produced through their fermentation by the intestinal microbiota, have long been thought to be the mediators of these benefits. In fact, propionate and butyrate were recently shown to activate intestinal gluconeogenesis, a function exerting metabolic benefits via its capacity of signaling to the brain by gastrointestinal nerves. Recently, succinate, the precursor of propionate in the bacterial metabolism, has also been shown to exert signaling properties, including the activation of intestinal gluconeogenesis.


Subject(s)
Brain/physiology , Energy Metabolism/physiology , Gastrointestinal Microbiome/physiology , Intestines/innervation , Intestines/physiology , Succinic Acid/metabolism , Animals , Homeostasis , Humans , Nerve Net/physiology , Signal Transduction/physiology
9.
Cell Metab ; 24(1): 151-7, 2016 07 12.
Article in English | MEDLINE | ID: mdl-27411015

ABSTRACT

Beneficial effects of dietary fiber on glucose and energy homeostasis have long been described, focusing mostly on the production of short-chain fatty acids by the gut commensal bacteria. However, bacterial fermentation of dietary fiber also produces large amounts of succinate and, to date, no study has focused on the role of succinate on host metabolism. Here, we fed mice a fiber-rich diet and found that succinate was the most abundant carboxylic acid in the cecum. Dietary succinate was identified as a substrate for intestinal gluconeogenesis (IGN), a process that improves glucose homeostasis. Accordingly, dietary succinate improved glucose and insulin tolerance in wild-type mice, but those effects were absent in mice deficient in IGN. Conventional mice colonized with the succinate producer Prevotella copri exhibited metabolic benefits, which could be related to succinate-activated IGN. Thus, microbiota-produced succinate is a previously unsuspected bacterial metabolite improving glycemic control through activation of IGN.


Subject(s)
Gluconeogenesis/drug effects , Glucose/metabolism , Homeostasis/drug effects , Intestinal Mucosa/metabolism , Microbiota , Succinic Acid/pharmacology , Animals , Cecum/drug effects , Cecum/metabolism , Feeding Behavior/drug effects , Genotype , Liver/drug effects , Liver/metabolism , Male , Mice, Inbred C57BL , Microbiota/drug effects , Oligosaccharides/pharmacology , Prevotella/drug effects , Prevotella/metabolism
10.
Cell ; 165(6): 1332-1345, 2016 Jun 02.
Article in English | MEDLINE | ID: mdl-27259147

ABSTRACT

A compelling set of links between the composition of the gut microbiota, the host diet, and host physiology has emerged. Do these links reflect cause-and-effect relationships, and what might be their mechanistic basis? A growing body of work implicates microbially produced metabolites as crucial executors of diet-based microbial influence on the host. Here, we will review data supporting the diverse functional roles carried out by a major class of bacterial metabolites, the short-chain fatty acids (SCFAs). SCFAs can directly activate G-coupled-receptors, inhibit histone deacetylases, and serve as energy substrates. They thus affect various physiological processes and may contribute to health and disease.


Subject(s)
Dietary Fiber/metabolism , Fatty Acids, Volatile/metabolism , Gastrointestinal Microbiome/physiology , Animals , Histones/metabolism , Humans , Intestines/immunology , Intestines/microbiology , Signal Transduction
11.
Mol Metab ; 5(4): 263-270, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27069866

ABSTRACT

OBJECTIVE: Insulin-like peptide 5 (INSL5) is a recently identified gut hormone that is produced predominantly by L-cells in the colon, but its function is unclear. We have previously shown that colonic expression of the gene for the L-cell hormone GLP-1 is high in mice that lack a microbiota and thus have energy-deprived colonocytes. Our aim was to investigate if energy deficiency also affected colonic Insl5 expression and to identify a potential role of INSL5. METHODS: We analyzed colonic Insl5 expression in germ-free (GF), conventionally raised (CONV-R), conventionalized (CONV-D) and antibiotic-treated mice, and also assessed the effect of dietary changes on colonic Insl5 expression. In addition, we characterized the metabolic phenotype of Insl5-/- mice. RESULTS: We showed that colonic Insl5 expression was higher in GF and antibiotic-treated mice than in CONV-R mice, whereas Insl5 expression in the brain was higher in CONV-R versus GF mice. We also observed that colonic Insl5 expression was suppressed by increasing the energy supply in GF mice by colonization or high-fat feeding. We did not observe any differences in food intake, gut transit or oral glucose tolerance between Insl5-/- and wild-type mice. However, we showed impaired intraperitoneal glucose tolerance in Insl5-/- mice. We also observed improved insulin tolerance and reduced hepatic glucose production in Insl5-/- mice. CONCLUSIONS: We have shown that colonic Insl5 expression is regulated by the gut microbiota and energy availability. We propose that INSL5 is a hormone that could play a role in promoting hepatic glucose production during periods of energy deprivation.

12.
Cell Metab ; 22(6): 971-82, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26552345

ABSTRACT

The gut microbiota plays an important role in human health by interacting with host diet, but there is substantial inter-individual variation in the response to diet. Here we compared the gut microbiota composition of healthy subjects who exhibited improved glucose metabolism following 3-day consumption of barley kernel-based bread (BKB) with those who responded least to this dietary intervention. The Prevotella/Bacteroides ratio was higher in responders than non-responders after BKB. Metagenomic analysis showed that the gut microbiota of responders was enriched in Prevotella copri and had increased potential to ferment complex polysaccharides after BKB. Finally, germ-free mice transplanted with microbiota from responder human donors exhibited improved glucose metabolism and increased abundance of Prevotella and liver glycogen content compared with germ-free mice that received non-responder microbiota. Our findings indicate that Prevotella plays a role in the BKB-induced improvement in glucose metabolism observed in certain individuals, potentially by promoting increased glycogen storage.


Subject(s)
Dietary Fiber/pharmacology , Glucose/metabolism , Microbiota/drug effects , Prevotella/growth & development , Aged , Animals , Bacteroides/genetics , Bacteroides/growth & development , Bacteroides/physiology , Blood Glucose/analysis , Cross-Over Studies , Feces/microbiology , Female , Glycogen/metabolism , Humans , Hydrogen/metabolism , Insulin/blood , Intestines/microbiology , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Middle Aged , Prevotella/genetics , Prevotella/physiology , RNA, Ribosomal, 16S/chemistry , RNA, Ribosomal, 16S/metabolism
13.
Med Sci (Paris) ; 31(2): 168-73, 2015 Feb.
Article in French | MEDLINE | ID: mdl-25744263

ABSTRACT

Since the XIX(th) century, the brain has been known for its role in regulating food intake (via the control of hunger sensation) and glucose homeostasis. Further interest has come from the discovery of gut hormones, which established a clear link between the gut and the brain in regulating glucose and energy homeostasis. The brain has two particular structures, the hypothalamus and the brainstem, which are sensitive to information coming either from peripheral organs or from the gut (via circulating hormones or nutrients) about the nutritional status of the organism. However, the efforts for a better understanding of these mechanisms have allowed to unveil a new gut-brain neural axis as a key regulator of the metabolic status of the organism. Certain nutrients control the hypothalamic homeostatic function via this axis. In this review, we describe how the gut is connected to the brain via different neural pathways, and how the interplay between these two organs drives the energy balance.


Subject(s)
Brain/physiology , Glucose/metabolism , Homeostasis/physiology , Intestines/physiology , Animals , Appetite Regulation/physiology , Autonomic Pathways/physiology , Blood Glucose/metabolism , Feeding Behavior/physiology , Gastrointestinal Hormones/physiology , Gluconeogenesis/physiology , Humans , Hunger/physiology , Hypothalamus/physiology , Intestines/innervation , Intestines/microbiology , Liver/metabolism , Microbiota , Satiety Response/physiology
14.
Ann Surg ; 262(6): 1006-15, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25575265

ABSTRACT

OBJECTIVE: To evaluate the role of bile routing modification on the beneficial effects of gastric bypass surgery on glucose and energy metabolism. BACKGROUND: Gastric bypass surgery (GBP) promotes early improvements in glucose and energy homeostasis in obese diabetic patients. A suggested mechanism associates a decrease in hepatic glucose production to an enhanced intestinal gluconeogenesis. Moreover, plasma bile acids are elevated after GBP and bile acids are inhibitors of gluconeogenesis. METHODS: In male Sprague-Dawley rats, we performed bile diversions from the bile duct to the midjejunum or the mid-ileum to match the modified bile delivery in the gut occurring in GBP. Body weight, food intake, glucose tolerance, insulin sensitivity, and food preference were analyzed. The expression of gluconeogenesis genes was evaluated in both the liver and the intestine. RESULTS: Bile diversions mimicking GBP promote an increase in plasma bile acids and a marked improvement in glucose control. Bile bioavailability modification is causal because a bile acid sequestrant suppresses the beneficial effects of bile diversions on glucose control. In agreement with the inhibitory role of bile acids on gluconeogenesis, bile diversions promote a blunting in hepatic glucose production, whereas intestinal gluconeogenesis is increased in the gut segments devoid of bile. In rats fed a high-fat-high-sucrose diet, bile diversions improve glucose control and dramatically decrease food intake because of an acquired disinterest in fatty food. CONCLUSIONS: This study shows that bile routing modification is a key mechanistic feature in the beneficial outcomes of GBP.


Subject(s)
Bile Acids and Salts/metabolism , Bile/physiology , Blood Glucose/metabolism , Energy Metabolism , Gastric Bypass , Animals , Biomarkers/metabolism , Food Preferences/physiology , Gene Expression Regulation , Gluconeogenesis/genetics , Homeostasis , Ileum/surgery , Insulin Resistance , Intestinal Mucosa/metabolism , Jejunum/surgery , Liver/metabolism , Male , Obesity/metabolism , Obesity/surgery , Rats , Rats, Sprague-Dawley
15.
Cell ; 156(1-2): 84-96, 2014 Jan 16.
Article in English | MEDLINE | ID: mdl-24412651

ABSTRACT

Soluble dietary fibers promote metabolic benefits on body weight and glucose control, but underlying mechanisms are poorly understood. Recent evidence indicates that intestinal gluconeogenesis (IGN) has beneficial effects on glucose and energy homeostasis. Here, we show that the short-chain fatty acids (SCFAs) propionate and butyrate, which are generated by fermentation of soluble fiber by the gut microbiota, activate IGN via complementary mechanisms. Butyrate activates IGN gene expression through a cAMP-dependent mechanism, while propionate, itself a substrate of IGN, activates IGN gene expression via a gut-brain neural circuit involving the fatty acid receptor FFAR3. The metabolic benefits on body weight and glucose control induced by SCFAs or dietary fiber in normal mice are absent in mice deficient for IGN, despite similar modifications in gut microbiota composition. Thus, the regulation of IGN is necessary for the metabolic benefits associated with SCFAs and soluble fiber.


Subject(s)
Gluconeogenesis , Intestinal Mucosa/metabolism , Intestines/innervation , Animals , Brain/metabolism , Dietary Fats/metabolism , Dietary Fiber/metabolism , Fatty Acids, Volatile/metabolism , Glucose/metabolism , Glucose-6-Phosphatase/genetics , Glucose-6-Phosphatase/metabolism , Homeostasis , Insulin Resistance , Mice , Microbiota , Obesity/metabolism , Oligosaccharides/metabolism , Rats
16.
Curr Opin Pharmacol ; 13(6): 959-63, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24095601

ABSTRACT

Mu-opioid receptors (MORs) are known to influence food intake at the brain level, through their involvement in the food reward system. MOR agonists stimulate food intake. On the other hand, MOR antagonists suppress food intake. MORs are also active in peripheral organs, especially in the small intestine where they control the gut motility. Recently, an indirect role in the control of food intake was ascribed to MORs in the extrinsic gastrointestinal neural system. MORs present in the neurons of the portal vein walls sense blood peptides released from the digestion of dietary protein. These peptides behave as MOR antagonists. Their MOR antagonist action initiates a gut-brain circuitry resulting in the induction of intestinal gluconeogenesis, a function controlling food intake. Thus, periportal MORs are a key mechanistic link in the satiety effect of protein-enriched diets.


Subject(s)
Portal Vein/innervation , Portal Vein/metabolism , Receptors, Opioid, mu/metabolism , Animals , Brain/metabolism , Eating/physiology , Gastrointestinal Tract/innervation , Gastrointestinal Tract/metabolism , Humans , Neurons/metabolism
17.
Eur J Neurosci ; 38(10): 3476-86, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24011250

ABSTRACT

The detection of glucose in the hepatoportal area is a simple but crucial peripheral cue initiating a nervous signal that ultimately leads to a wide array of metabolic and behavioural responses, such as decreased food intake, tighter control of glucose homeostasis, or appearance of food preference. This signal has been suggested to mediate the effects of high-protein diets, as opposed to high-fat/high-sucrose diets. Nevertheless, the central targets of the signal originating from the hepatoportal area remain largely undocumented. Using immunohistochemistry on the brain of male rats, we show here that portal glucose increases c-Fos expression in the brainstem, in the hypothalamus (in particular in neurons expressing pro-opiomelanocortin) and also in olfactory and other limbic and cortical areas, including those functionally implicated in reward (Experiment 1). In similar postabsorptive conditions, a high-protein diet induced similar effects in the hypothalamus and the granular cells of the main olfactory bulb, whereas the high-fat/high-sucrose diet actually reduced the basal expression of c-Fos in cortical layers. Both diets also decreased the number of neurons expressing c-Fos in the amygdala and gustatory areas (Experiment 2). Altogether, these findings suggest that the peripheral signal primed by portal glucose sensing may influence behavioural adaptation such as food preference via a network including the olfactory pathway, central amygdala, nucleus accumbens and orbitofrontal cortex, in addition to satiety and metabolic effects primarily implicating the hypothalamic response.


Subject(s)
Cerebral Cortex/metabolism , Glucose/physiology , Hypothalamus/metabolism , Olfactory Bulb/metabolism , Portal System/physiology , Reward , Animals , Brain Stem/metabolism , Brain Stem/physiology , Cerebral Cortex/physiology , Eating/physiology , Hypothalamus/physiology , Male , Olfactory Bulb/physiology , Rats , Rats, Sprague-Dawley
19.
Cell ; 150(2): 377-88, 2012 Jul 20.
Article in English | MEDLINE | ID: mdl-22771138

ABSTRACT

Intestinal gluconeogenesis is involved in the control of food intake. We show that mu-opioid receptors (MORs) present in nerves in the portal vein walls respond to peptides to regulate a gut-brain neural circuit that controls intestinal gluconeogenesis and satiety. In vitro, peptides and protein digests behave as MOR antagonists in competition experiments. In vivo, they stimulate MOR-dependent induction of intestinal gluconeogenesis via activation of brain areas receiving inputs from gastrointestinal ascending nerves. MOR-knockout mice do not carry out intestinal gluconeogenesis in response to peptides and are insensitive to the satiety effect induced by protein-enriched diets. Portal infusions of MOR modulators have no effect on food intake in mice deficient for intestinal gluconeogenesis. Thus, the regulation of portal MORs by peptides triggering signals to and from the brain to induce intestinal gluconeogenesis are links in the satiety phenomenon associated with alimentary protein assimilation.


Subject(s)
Dietary Proteins/metabolism , Eating , Gluconeogenesis , Receptors, Opioid, mu/metabolism , Satiety Response , Animals , Brain/metabolism , Humans , Intestinal Mucosa/metabolism , Male , Mice , Mice, Knockout , Rats , Rats, Sprague-Dawley , Receptors, Opioid, mu/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...