Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 262
Filter
1.
Int J Mol Sci ; 25(11)2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38892414

ABSTRACT

Berberine (BBR) is used to treat cancer, inflammatory conditions, and so on. But the side effects of BBR causing constipation should not be ignored. In clinical application, the combination of Amomum villosum Lour. (AVL) and BBR can relieve it. However, the effective ingredients and molecular mechanism of AVL in relieving constipation are not clear. A small intestine propulsion experiment was conducted in constipated mice to screen active ingredients of AVL. We further confirmed the molecular mechanism of action of the active ingredient on BBR-induced constipation. Quercetin (QR) was found to be the effective ingredient of AVL in terms of relieving constipation. QR can efficiently regulate the microbiota in mice suffering from constipation. Moreover, QR significantly raised the levels of substance P and motilin while lowering those of 5-hydroxytryptamine and vasoactive intestinal peptide; furthermore, it also increased the protein expression levels of calmodulin, myosin light-chain kinase, and myosin light chain. The use of QR in combination with BBR has an adverse effect-reducing efficacy. The study provides new ideas and possibilities for the treatment of constipation induced by BBR.


Subject(s)
Berberine , Constipation , Gastrointestinal Microbiome , Quercetin , Animals , Berberine/pharmacology , Berberine/therapeutic use , Quercetin/pharmacology , Constipation/drug therapy , Constipation/chemically induced , Gastrointestinal Microbiome/drug effects , Mice , Male , Disease Models, Animal , Motilin/metabolism
2.
Cell Tissue Res ; 397(1): 61-76, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38727755

ABSTRACT

Motilin (MLN) is a peptide hormone originally isolated from the mucosa of the porcine intestine. Its orthologs have been identified in various vertebrates. Although MLN regulates gastrointestinal motility in tetrapods from amphibians to mammals, recent studies indicate that MLN is not involved in the regulation of isolated intestinal motility in zebrafish, at least in vitro. To determine the unknown function of MLN in teleosts, we examined the expression of MLN and the MLN receptor (MLNR) at the cellular level in Japanese medaka (Oryzias latipes). Quantitative PCR revealed that mln mRNA was limitedly expressed in the gut, whereas mlnr mRNA was not detected in the gut but was expressed in the brain and kidney. By in situ hybridization and immunohistochemistry, mlnr mRNA was detected in the dopaminergic neurons of the area postrema in the brain and the noradrenaline-producing cells in the interrenal gland of the kidney. Furthermore, we observed efferent projections of mlnr-expressing dopaminergic neurons in the lobus vagi (XL) and nucleus motorius nervi vagi (NXm) of the medulla oblongata by establishing a transgenic medaka expressing the enhanced green fluorescence protein driven by the mlnr promoter. The expression of dopamine receptor mRNAs in the XL and cholinergic neurons in NXm was confirmed by in situ hybridization. These results indicate novel sites of MLN activity other than the gastrointestinal tract. MLN may exert central and peripheral actions through the regulation of catecholamine release in medaka.


Subject(s)
Motilin , Oryzias , Receptors, Gastrointestinal Hormone , Animals , Oryzias/metabolism , Oryzias/genetics , Receptors, Gastrointestinal Hormone/metabolism , Receptors, Gastrointestinal Hormone/genetics , Motilin/metabolism , Receptors, Neuropeptide/metabolism , Receptors, Neuropeptide/genetics , RNA, Messenger/metabolism , RNA, Messenger/genetics , Animals, Genetically Modified , Dopaminergic Neurons/metabolism , Brain/metabolism
3.
Benef Microbes ; 15(3): 311-329, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38688519

ABSTRACT

Probiotics exert beneficial effects by regulating the intestinal microbiota, metabolism, immune function and other ways of their host. Patients with constipation, a common gastrointestinal disorder, experience disturbances in their intestinal microbiota. In the present study, we investigated the effectiveness of two microbial ecological agents (postbiotic extract PE0401 and a combination of postbiotic extract PE0401 and Lacticaseibacillus paracasei CCFM 2711) in regulating the makeup of the intestinal microbiota and alleviating loperamide hydrochloride-induced constipation in mice. We also preliminarily explored the mechanism underlying their effects. Both microbial ecological agents increased the abundance of the beneficial bacteria Lactobacilli and Bifidobacterium after administration and were able to relieve constipation. However, the degree of improvement in constipation symptoms varied depending on the makeup of the supplement. The postbiotic extract PE0401 increased peristalsis time and improved faecal properties throughout the intestinal tract of the host. PE0401 relieved constipation, possibly by modulating the levels of the constipation-related gastrointestinal regulatory transmitters mouse motilin, mouse vasoactive intestinal peptide, and 5-hydoxytryptamine in the intestinal tract of the host and by increasing the levels of the short-chain fatty acids (SCFAs) acetic acid, propionic acid, and isovaleric acid. It also increased the relative abundance of Lactobacillus and Bifidobacterium and reduced that of Faecalibaculum, Mucispirillum, Staphylococcus, and Lachnoclostridium, which are among the beneficial microbiota in the host intestine. Furthermore, PE0401 decreased the levels of constipation-induced host inflammatory factors. Therefore, the two microbial ecological agents can regulate the intestinal microbiota of constipation mice, and PE0401 has a stronger ability to relieve constipation.


Subject(s)
Constipation , Fatty Acids, Volatile , Gastrointestinal Microbiome , Loperamide , Probiotics , Animals , Loperamide/adverse effects , Constipation/drug therapy , Constipation/chemically induced , Constipation/microbiology , Gastrointestinal Microbiome/drug effects , Fatty Acids, Volatile/metabolism , Mice , Probiotics/administration & dosage , Probiotics/pharmacology , Probiotics/therapeutic use , Male , Bifidobacterium , Lacticaseibacillus paracasei , Disease Models, Animal , Lactobacillus , Motilin/metabolism , Feces/microbiology , Feces/chemistry , Vasoactive Intestinal Peptide/metabolism
4.
Biomolecules ; 14(4)2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38672450

ABSTRACT

Motilin is a gastrointestinal hormone that is mainly produced in the duodenum of mammals, and it is responsible for regulating appetite. However, the role and expression of motilin are poorly understood during starvation and the weaning stage, which is of great importance in the seeding cultivation of fish. In this study, the sequences of Yangtze sturgeon (Acipenser dabryanus Motilin (AdMotilin)) motilin receptor (AdMotilinR) were cloned and characterized. The results of tissue expression showed that by contrast with mammals, AdMotilin mRNA was richly expressed in the brain, whereas AdMotilinR was highly expressed in the stomach, duodenum, and brain. Weaning from a natural diet of T. Limnodrilus to commercial feed significantly promoted the expression of AdMotilin in the brain during the period from day 1 to day 10, and after re-feeding with T. Limnodrilus the change in expression of AdMotilin was partially reversed. Similarly, it was revealed that fasting increased the expression of AdMotilin in the brain (3 h, 6 h) and duodenum (3 h), and the expression of AdMotilinR in the brain (1 h) in a time-dependent manner. Furthermore, it was observed that peripheral injection of motilin-NH2 increased food intake and the filling index of the digestive tract in the Yangtze sturgeon, which was accompanied by the changes of AdMotilinR and appetite factors expression in the brain (POMC, CART, AGRP, NPY and CCK) and stomach (CCK). These results indicate that motilin acts as an indicator of nutritional status, and also serves as a novel orexigenic factor that stimulates food intake in Acipenser dabryanus. This study lays a strong foundation for the application of motilin as a biomarker in the estimation of hunger in juvenile Acipenser dabryanu during the weaning phase, and enhances the understanding of the role of motilin as a novel regulator of feeding in fish.


Subject(s)
Feeding Behavior , Fishes , Motilin , Animals , Brain/metabolism , Fish Proteins/metabolism , Fishes/metabolism , Fishes/genetics , Fishes/physiology , Motilin/genetics , Motilin/metabolism , Motilin/pharmacology , Receptors, Gastrointestinal Hormone/metabolism , Receptors, Gastrointestinal Hormone/genetics , Receptors, Neuropeptide/metabolism , Receptors, Neuropeptide/genetics
5.
Food Funct ; 15(4): 2221-2233, 2024 Feb 19.
Article in English | MEDLINE | ID: mdl-38318756

ABSTRACT

Motilin is an important hormonal regulator in the migrating motor complex (MMC). Free fatty acid receptor-1 (FFAR1, also known as GPR40) has been reported to stimulate motilin release in human duodenal organoids. However, how FFAR1 regulates gastric motility in vivo is unclear. This study investigated the role of FFAR1 in the regulation of gastric contractions and its possible mechanism of action using Suncus murinus. Firstly, intragastric administration of oleic acid (C18:1, OA), a natural ligand for FFAR1, stimulated phase II-like contractions, followed by phase III-like contractions in the fasted state, and the gastric emptying rate was accelerated. The administration of GW1100, an FFAR1 antagonist, inhibited the effects of OA-induced gastric contractions. Intravenous infusion of a ghrelin receptor antagonist (DLS) or serotonin 4 (5-HT4) receptor antagonist (GR125487) inhibited phase II-like contractions and prolonged the onset of phase III-like contractions induced by OA. MA-2029, a motilin receptor antagonist, delayed the occurrence of phase III-like contractions. In vagotomized suncus, OA did not induce phase II-like contractions. In addition, OA promoted gastric emptying through a vagal pathway during the postprandial period. However, OA did not directly act on the gastric body to induce contractions in vitro. In summary, this study indicates that ghrelin, motilin, 5-HT, and the vagus nerve are involved in the role of FFAR1 regulating MMC. Our findings provide novel evidence for the involvement of nutritional factors in the regulation of gastric motility.


Subject(s)
Fatty Acids, Nonesterified , Gastrointestinal Motility , Humans , Animals , Fatty Acids, Nonesterified/pharmacology , Motilin/metabolism , Motilin/pharmacology , Myoelectric Complex, Migrating/physiology , Stomach/physiology , Shrews/metabolism
6.
Sci Adv ; 9(11): eade9020, 2023 03 17.
Article in English | MEDLINE | ID: mdl-36921049

ABSTRACT

Motilin is an endogenous peptide hormone almost exclusively expressed in the human gastrointestinal (GI) tract. It activates the motilin receptor (MTLR), a class A G protein-coupled receptor (GPCR), and stimulates GI motility. To our knowledge, MTLR is the first GPCR reported to be activated by macrolide antibiotics, such as erythromycin. It has attracted extensive attention as a potential drug target for GI disorders. We report two structures of Gq-coupled human MTLR bound to motilin and erythromycin. Our structures reveal the recognition mechanism of both ligands and explain the specificity of motilin and ghrelin, a related gut peptide hormone, for their respective receptors. These structures also provide the basis for understanding the different recognition modes of erythromycin by MTLR and ribosome. These findings provide a framework for understanding the physiological regulation of MTLR and guiding drug design targeting MTLR for the treatment of GI motility disorders.


Subject(s)
Motilin , Receptors, Gastrointestinal Hormone , Humans , Motilin/metabolism , Erythromycin/pharmacology , Erythromycin/metabolism , Receptors, Gastrointestinal Hormone/chemistry , Receptors, Gastrointestinal Hormone/metabolism , Receptors, Neuropeptide/metabolism
7.
Comput Math Methods Med ; 2022: 5169892, 2022.
Article in English | MEDLINE | ID: mdl-35799630

ABSTRACT

Functional dyspepsia (FD) is a common digestive system disease, and probiotics in the treatment of FD have a good curative effect. Patients with gastrointestinal diseases often show a poor response to traditional drug treatments and suffer from adverse reactions. Kvass can be used as a functional drink without side effects to improve the symptoms of FD patients. The results showed that compared with those of the model group, the body weight and food intake of the treatment group were significantly increased (P < 0.05), and the gastric residual rate of the treatment group was significantly decreased (P < 0.05); the amount of pepsin in the treatment group was significantly higher than that in the model group (P < 0.05); a high dose of Kvass could increase the contents of ghrelin, motilin (MTL), and gastrin (GAS) in the plasma and decrease the contents of vasoactive intestinal peptide (VIP) in the plasma; the contents of ghrelin, MTL, and GAS in the gastric antrum were also increased in the high-dose group. Kvass beverage can significantly improve the gastrointestinal function of rats, which may be because it can improve the contents of ghrelin, MTL, GAS, and VIP in both the serum and gastric antrum by regulating the expression of short-chain fatty acids in the colon.


Subject(s)
Dyspepsia , Animals , Dyspepsia/drug therapy , Gastrointestinal Motility/physiology , Ghrelin , Motilin/metabolism , Motilin/pharmacology , Rats , Stomach/physiology , Vasoactive Intestinal Peptide/metabolism , Vasoactive Intestinal Peptide/pharmacology
8.
Pharmacol Res Perspect ; 10(2): e00900, 2022 04.
Article in English | MEDLINE | ID: mdl-35191209

ABSTRACT

The gastrointestinal (GI) hormone motilin helps control human stomach movements during hunger and promotes hunger. Although widely present among mammals, it is generally accepted that in rodents the genes for motilin and/or its receptor have undergone pseudonymization, so exogenous motilin cannot function. However, several publications describe functions of low concentrations of motilin, usually within the GI tract and CNS of mice, rats, and guinea pigs. These animals were from institute-held stocks, simply described with stock names (e.g., "Sprague-Dawley") or were inbred strains. It is speculated that variation in source/type of animal introduces genetic variations to promote motilin-sensitive pathways. Perhaps, in some populations, motilin receptors exist, or a different functionally-active receptor has a good affinity for motilin (indicating evolutionary pressures to retain motilin functions). The ghrelin receptor has the closest sequence homology, yet in non-rodents the receptors have a poor affinity for each other's cognate ligand. In rodents, ghrelin may substitute for certain GI functions of motilin, but no good evidence suggests rodent ghrelin receptors are highly responsive to motilin. It remains unknown if motilin has functional relationships with additional bioactive molecules formed from the ghrelin and motilin genes, or if a 5-TM motilin receptor has influence in rodents (e.g., to dimerize with GPCRs and create different pharmacological profiles). Is the absence/presence of responses to motilin in rodents' characteristic for systems undergoing gene pseudonymization? What are the consequences of rodent supplier-dependent variations in motilin sensitivity (or other ligands for receptors undergoing pseudonymization) on gross physiological functions? These are important questions for understanding animal variation.


Subject(s)
Gastrointestinal Tract/physiology , Motilin/metabolism , Receptors, Gastrointestinal Hormone/metabolism , Receptors, Neuropeptide/metabolism , Animals , Genetic Variation , Ghrelin/metabolism , Guinea Pigs , Humans , Mice , Rats , Receptors, Ghrelin/metabolism , Rodentia , Species Specificity
9.
Mol Metab ; 54: 101356, 2021 12.
Article in English | MEDLINE | ID: mdl-34662713

ABSTRACT

OBJECTIVE: Motilin is a proximal small intestinal hormone with roles in gastrointestinal motility, gallbladder emptying, and hunger initiation. In vivo motilin release is stimulated by fats, bile, and duodenal acidification but the underlying molecular mechanisms of motilin secretion remain poorly understood. This study aimed to establish the key signaling pathways involved in the regulation of secretion from human motilin-expressing M-cells. METHODS: Human duodenal organoids were CRISPR-Cas9 modified to express the fluorescent protein Venus or the Ca2+ sensor GCaMP7s under control of the endogenous motilin promoter. This enabled the identification and purification of M-cells for bulk RNA sequencing, peptidomics, calcium imaging, and electrophysiology. Motilin secretion from 2D organoid-derived cultures was measured by liquid chromatography-tandem mass spectrometry (LC-MS/MS), in parallel with other gut hormones. RESULTS: Human duodenal M-cells synthesize active forms of motilin and acyl-ghrelin in organoid culture, and also co-express cholecystokinin (CCK). Activation of the bile acid receptor GPBAR1 stimulated a 3.4-fold increase in motilin secretion and increased action potential firing. Agonists of the long-chain fatty acid receptor FFA1 and monoacylglycerol receptor GPR119 stimulated secretion by 2.4-fold and 1.5-fold, respectively. Acidification (pH 5.0) was a potent stimulus of M-cell calcium elevation and electrical activity, an effect attributable to acid-sensing ion channels, and a modest inducer of motilin release. CONCLUSIONS: This study presents the first in-depth transcriptomic and functional characterization of human duodenal motilin-expressing cells. We identify several receptors important for the postprandial and interdigestive regulation of motilin release.


Subject(s)
Bile/metabolism , Duodenum/metabolism , Fatty Acids, Nonesterified/metabolism , Motilin/metabolism , Organoids/metabolism , Cells, Cultured , Humans , Hydrogen-Ion Concentration
10.
Front Endocrinol (Lausanne) ; 12: 700884, 2021.
Article in English | MEDLINE | ID: mdl-34497583

ABSTRACT

Motilin, produced in endocrine cells in the mucosa of the upper intestine, is an important regulator of gastrointestinal (GI) motility and mediates the phase III of interdigestive migrating motor complex (MMC) in the stomach of humans, dogs and house musk shrews through the specific motilin receptor (MLN-R). Motilin-induced MMC contributes to the maintenance of normal GI functions and transmits a hunger signal from the stomach to the brain. Motilin has been identified in various mammals, but the physiological roles of motilin in regulating GI motility in these mammals are well not understood due to inconsistencies between studies conducted on different species using a range of experimental conditions. Motilin orthologs have been identified in non-mammalian vertebrates, and the sequence of avian motilin is relatively close to that of mammals, but reptile, amphibian and fish motilins show distinctive different sequences. The MLN-R has also been identified in mammals and non-mammalian vertebrates, and can be divided into two main groups: mammal/bird/reptile/amphibian clade and fish clade. Almost 50 years have passed since discovery of motilin, here we reviewed the structure, distribution, receptor and the GI motility regulatory function of motilin in vertebrates from fish to mammals.


Subject(s)
Gastrointestinal Motility , Motilin/metabolism , Muscle Contraction , Receptors, Gastrointestinal Hormone/metabolism , Receptors, Neuropeptide/metabolism , Animals , Humans
11.
Peptides ; 143: 170574, 2021 09.
Article in English | MEDLINE | ID: mdl-34082070

ABSTRACT

BACKGROUND: Motilin's role in the regulation of vascular tone and hemodynamic besides gastrointestinal motility is concerned. This study aimed to investigate the expression of motilin receptors in gastrointestinal arteries and motilin-induced relaxation. MATERIAL AND METHODS: The expression of motilin receptors in the left gastric artery (LGA), superior mesenteric artery (SMA), and inferior mesenteric artery (IMA) of adult dogs (1.5-5 years old) were analyzed by immunochemistry, RT-PCR, and western blotting. Motilin's effects on the gastrointestinal arteries were evaluated in a multi-wire myograph system. RESULTS: Immunohistochemical staining showed that motilin receptor was expressed on the membranes of endothelial cells with the fluorescence intensity LGA > SMA > IMA (P < 0.01). The motilin receptor's mRNA and protein expression levels shared the same distribution patterns as it in fluorescence intensity (P < 0.01). In isolated LGA preparations precontracted with U46619 (a thromboxaneA2 analog), motilin induced a concentration-dependent relaxation, and the EC50 was 8.8 × 10-8 ± 0.9 × 10-8 M. Motilin-induced relaxation on the three arteries also shared the same pattern as it in fluorescence intensity (P < 0.01) and inhibited by denuded-endothelium and GM-109 (a motilin receptor antagonist) but not by atropine (a muscarinic receptor antagonist). CONCLUSIONS: Motilin receptors are expressed differentially on the membranes of endothelial cells in dog gastrointestinal arteries with a significantly high expression in the LGA. Motilin-induced relaxation is endothelium- and motilin receptor-dependent. The motilin receptor expressed on the endothelial cell membrane of the LGA is the molecular basis for motilin regulating gastric blood flow under physiological conditions in dogs.


Subject(s)
Arteries/metabolism , Endothelium, Vascular/metabolism , Motilin/metabolism , Receptors, Gastrointestinal Hormone/genetics , Receptors, Neuropeptide/genetics , Animals , Dogs , Female , Gastrointestinal Tract/blood supply , Gene Expression Regulation , Male , Receptors, Gastrointestinal Hormone/metabolism , Receptors, Neuropeptide/metabolism
12.
J Cell Mol Med ; 25(8): 3754-3764, 2021 04.
Article in English | MEDLINE | ID: mdl-33629528

ABSTRACT

In this study, we aimed to investigate the role of circORC2 in modulating miR-19a and its downstream signalling during the pathogenesis of STC. In this study, three groups of patients, that is healthy control (HC) group, normal transit constipation (NTC) group (N = 42) and slow transit constipation (STC) group, were, respectively, recruited. RT-PCR and Western blot analysis were exploited to investigate the changes in the expression levels of miR-19a and circORC2 in these patients, so as to establish a circORC2/miR-19a signalling pathway. The basic information of the patients showed no significant differences among different patient groups. Compared with the HC group, concentrations of neurotensin (NST) and motilin (MLN) were both significantly reduced in the NTC and STC groups, especially in the STC group. Also, miR-19a level was highest, whereas circORC2 level was lowest in the STC group. Furthermore, circORC2 was validated to sponge the expression of miR-19a, and the transfection of circORC2 reduced the expression of miR-19a. Meanwhile, MLN and NST mRNAs were both targeted by miR-19a, and the transfection of circORC2 dramatically up-regulated the expression of MLN and NST. On the contrary, the transfection of circORC2 siRNA into SMCs and VSMCs exhibited the opposite effect of circORC2. Collectively, the results of this study established a regulatory relationship among circORC2, miR-19a and neurotensin/motilin, which indicated that the overexpression of circORC2 could up-regulate the levels of neurotensin and motilin, thus exerting a beneficial effect during the treatment of STC.


Subject(s)
Biomarkers/metabolism , Constipation/pathology , Gene Expression Regulation , MicroRNAs/genetics , Motilin/metabolism , Neurotensin/metabolism , RNA, Circular/genetics , Aged , Apoptosis , Case-Control Studies , Cell Proliferation , Cells, Cultured , Constipation/genetics , Constipation/metabolism , Female , Humans , Male , Motilin/genetics , Neurotensin/genetics , Origin Recognition Complex , Prognosis
13.
J Med Food ; 22(12): 1208-1221, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31621475

ABSTRACT

This study determined the ameliorative effects of the novel microorganism, Lactobacillus plantarum CQPC02 (LP-CQPC02), fermented in soybean milk, on loperamide-induced constipation in Kunming mice. High-performance liquid chromatography revealed that LP-CQPC02-fermented soybean milk (LP-CQPC02-FSM) had six types of soybean isoflavones, whereas Lactobacillus bulgaricus-fermented soybean milk (LB-FSM) and unfermented soybean milk (U-FSM) only had five types of soybean isoflavones. LP-CQPC02-FSM also contained more total and active soybean isoflavones than LB-FSM and U-FSM. Results from mouse experiments showed that the defecation factors (quantity, fecal weight and water content, gastrointestinal transit ability, and time to first black stool) in the LP-CQPC02-FSM-treated mice were better than those in the LB-FSM- and U-FSM-treated mice. The serum and small intestinal tissue experiments showed that soybean milk increased the motilin, gastrin, endothelin, acetylcholinesterase, substance P, vasoactive intestinal peptide, and glutathione levels and decreased the somatostatin, myeloperoxidase, nitric oxide, and malondialdehyde levels compared with the constipated mice in the control group. The LP-CQPC02-FSM also showed better effects than those of LB-FSM and U-FSM. Further results showed that LP-CQPC02-FSM upregulated cuprozinc-superoxide dismutase (Cu/Zn-SOD), manganese superoxide dismutase (Mn-SOD), catalase (CAT), c-Kit, stem cell factor (SCF), glial cell-derived neurotrophic factor (GDNF), neuronal nitric oxide synthase (nNOS), endothelial nitric oxide synthase (eNOS), and aquaporin-9 (AQP9) and downregulated the expression levels of transient receptor potential cation channel subfamily V member 1 (TRPV1), inducible nitric oxide synthase (iNOS), and aquaporin-3 (AQP3) in the constipated mice. LP-CQPC02-FSM increased the Bacteroides and Akkermansia abundances and decreased the Firmicutes abundance in the feces of the constipated mice and decreased the Firmicutes/Bacteroides ratio. This study confirmed that LP-CQPC02-FSM partially reversed constipation in mice.


Subject(s)
Constipation/therapy , Fermentation , Glycine max/metabolism , Lactobacillus plantarum/metabolism , Loperamide/adverse effects , Milk/metabolism , Soy Foods , Acetylcholinesterase/metabolism , Animals , Aquaporin 3/metabolism , Aquaporins , Catalase/metabolism , Constipation/chemically induced , Disease Models, Animal , Endothelins/metabolism , Feces/microbiology , Female , Gastrins/metabolism , Gastrointestinal Transit , Intestinal Mucosa/metabolism , Intestines/pathology , Isoflavones , Lactobacillus plantarum/isolation & purification , Malondialdehyde/metabolism , Mice , Motilin/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , Proto-Oncogene Proteins c-kit , Stem Cell Factor/metabolism , Superoxide Dismutase/metabolism , Superoxide Dismutase-1/metabolism , TRPV Cation Channels/metabolism
14.
Neurogastroenterol Motil ; 31(4): e13546, 2019 04.
Article in English | MEDLINE | ID: mdl-30740834

ABSTRACT

There have been many recent advances in the understanding of various aspects of the physiology of gastric motility and gastric emptying. Earlier studies had discovered the remarkable ability of the stomach to regulate the timing and rate of emptying of ingested food constituents and the underlying motor activity. Recent studies have shown that two parallel neural circuits, the gastric inhibitory vagal motor circuit (GIVMC) and the gastric excitatory vagal motor circuit (GEVMC), mediate gastric inhibition and excitation and therefore the rate of gastric emptying. The GIVMC includes preganglionic cholinergic neurons in the DMV and the postganglionic inhibitory neurons in the myenteric plexus that act by releasing nitric oxide, ATP, and peptide VIP. The GEVMC includes distinct gastric excitatory preganglionic cholinergic neurons in the DMV and postganglionic excitatory cholinergic neurons in the myenteric plexus. Smooth muscle is the final target of these circuits. The role of the intramuscular interstitial cells of Cajal in neuromuscular transmission remains debatable. The two motor circuits are differentially regulated by different sets of neurons in the NTS and vagal afferents. In the digestive period, many hormones including cholecystokinin and GLP-1 inhibit gastric emptying via the GIVMC, and in the inter-digestive period, hormones ghrelin and motilin hasten gastric emptying by stimulating the GEVMC. The GIVMC and GEVMC are also connected to anorexigenic and orexigenic neural pathways, respectively. Identification of the control circuits of gastric emptying may provide better delineation of the pathophysiology of abnormal gastric emptying and its relationship to satiety signals and food intake.


Subject(s)
Enteric Nervous System/physiology , Gastric Emptying/physiology , Neurons/physiology , Animals , Gastrointestinal Motility/physiology , Ghrelin/metabolism , Humans , Motilin/metabolism
15.
Gen Comp Endocrinol ; 277: 38-48, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30771290

ABSTRACT

Motilin (MLN), an interdigestive hormone secreted by endocrine cells of the intestinal mucosa, binds to a G protein-coupled receptor to exert its biological function of regulating gastrointestinal motility. In the present study, we identified the prepromotilin and mln receptor (mlnr) from the spotted sea bass, Lateolabrax maculatus. Mln consisted of an ORF of 336 nucleotides encoding 111 amino acids. The precursor protein contained a 17-amino-acid mature peptide. Mlnr had an ORF of 1089 bp encoding a protein of 362 amino acids. Seven transmembrane domains were predicted with TMHMM analysis. The phylogenetic analysis of mln and mlnr showed that they fell into the same clade with respective counterpart of selected fishes before clustering with other detected vertebrates. Both mln and mlnr genes were highly expressed in intestine of spotted sea bass using quantitative real-time PCR. In situ hybridization indicated that mln and mlnr mRNA were both localized in the lamina propria and the epithelial cell of intestinal villus. The expressions of both genes were regulated under short-term starvation in a time-dependent manner. In vitro experiments indicated that the expressions of ghrelin (ghrl), gastrin (gas) and cholecystokinin (cck) were enhanced by MLN after 3-h treatment, but the effect was absent after 6 or 12-h incubation. Taken together, the MLN and its receptor might play important roles in regulating intestinal motility in spotted sea bass.


Subject(s)
Bass/genetics , Gene Expression Profiling , Motilin/genetics , Receptors, Gastrointestinal Hormone/genetics , Receptors, Neuropeptide/genetics , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , Fasting , Gene Expression Regulation , Intestines/cytology , Molecular Docking Simulation , Motilin/chemistry , Motilin/metabolism , Phylogeny , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Gastrointestinal Hormone/chemistry , Receptors, Gastrointestinal Hormone/metabolism , Receptors, Neuropeptide/chemistry , Receptors, Neuropeptide/metabolism , Sequence Analysis, DNA
16.
PLoS One ; 14(2): e0205939, 2019.
Article in English | MEDLINE | ID: mdl-30789939

ABSTRACT

Motilin is a gastrointestinal peptide hormone that stimulates gastrointestinal motility. Motilin is produced primarily in the duodenum and jejunum. Motilin receptors (MTLRs) are G protein-coupled receptors that may represent a clinically useful pharmacological target as they can be activated by erythromycin. The functions of motilin are highly species-dependent and remain poorly understood. As a functional motilin system is absent in rodents such as rats and mice, these species are not commonly used for basic studies. In this study, we examine the usefulness of human MTLR-overexpressing transgenic (hMTLR-Tg) mice by identifying the mechanisms of the gastric motor response to human motilin and erythromycin. The distribution of hMTLR was examined immunohistochemically in male wild-type (WT) and hMTLR-Tg mice. The contractile response of gastric strips was measured isometrically in an organ bath, while gastric emptying was determined using phenol red. hMTLR expression was abundant in the gastric smooth muscle layer. Interestingly, higher levels of hMTLR expression were observed in the myenteric plexus of hMTLR-Tg mice but not WT mice. hMTLR was not co-localized with vesicular acetylcholine transporter, a marker of cholinergic neurons in the myenteric plexus. Treatment with human motilin and erythromycin caused concentration-dependent contraction of gastric strips obtained from hMTLR-Tg mice but not from WT mice. The contractile response to human motilin and erythromycin in hMTLR-Tg mice was affected by neither atropine nor tetrodotoxin and was totally absent in Ca2+-free conditions. Furthermore, intraperitoneal injection of erythromycin significantly promoted gastric emptying in hMTLR-Tg mice but not in WT mice. Human motilin and erythromycin stimulate gastric smooth muscle contraction in hMTLR-Tg mice. This action is mediated by direct contraction of smooth muscle via the influx of extracellular Ca2+. Thus, hMTLR-Tg mice may be useful for the evaluation of MTLR agonists as gastric prokinetic agents.


Subject(s)
Erythromycin/metabolism , Gastrointestinal Motility/physiology , Motilin/metabolism , Receptors, Gastrointestinal Hormone/metabolism , Receptors, Neuropeptide/metabolism , Animals , Calcium/metabolism , Cations, Divalent/metabolism , Cholinergic Neurons/cytology , Cholinergic Neurons/metabolism , Gene Expression , Humans , Male , Mice, Inbred C57BL , Mice, Transgenic , Muscle Contraction/physiology , Muscle, Smooth/cytology , Muscle, Smooth/metabolism , Myenteric Plexus/cytology , Myenteric Plexus/metabolism , Receptors, Gastrointestinal Hormone/genetics , Receptors, Neuropeptide/genetics , Stomach/cytology , Stomach/physiology , Tissue Culture Techniques
17.
Nat Rev Endocrinol ; 15(4): 238-250, 2019 04.
Article in English | MEDLINE | ID: mdl-30675023

ABSTRACT

After the discovery of motilin in 1972, motilin and the motilin receptor were studied intensely for their role in the control of gastrointestinal motility and as targets for treating hypomotility disorders. The genetic revolution - with the use of knockout models - sparked novel insights into the role of multiple peptides but contributed to a decline in interest in motilin, as this peptide and its receptor exist only as pseudogenes in rodents. The past 5 years have seen a major surge in interest in motilin, as a series of studies have shown its relevance in the control of hunger and regulation of food intake in humans in both health and disease. Luminal stimuli, such as bitter tastants, have been identified as modulators of motilin release, with effects on hunger and food intake. The current state of knowledge and potential implications for therapy are summarized in this Review.


Subject(s)
Gastric Emptying/physiology , Gastrointestinal Motility/physiology , Hunger/physiology , Motilin/metabolism , Receptors, Gastrointestinal Hormone/antagonists & inhibitors , Receptors, Neuropeptide/antagonists & inhibitors , Animals , Dogs , Eating/drug effects , Gastric Emptying/drug effects , Gastrointestinal Motility/drug effects , Humans , Hunger/drug effects , Mice , Receptors, Gastrointestinal Hormone/metabolism , Receptors, Neuropeptide/metabolism , Sensitivity and Specificity
18.
J Agric Food Chem ; 66(16): 4106-4117, 2018 Apr 25.
Article in English | MEDLINE | ID: mdl-29627986

ABSTRACT

This study was to probe the effects of bacterial cellulose (BC) on diphenoxylate-induced constipation in rats. Administration with BC at 500 mg/kg of body weight in diphenoxylate-induced constipation rats distinctly improved the carmine propulsion rate (83.5 ± 5.2%), shortened the defecating time of the first red feces (249.0 ± 23.3 min), and increased the weight of carmine red feces within 5 h (2.7 ± 1.3 g). The levels of aquaporins (AQP-2, AQP-3, and AQP-4) and inhibitory neurotransmitters (nitric oxide, nitric oxide synthetase, vasoactive intestinal peptide, and arginine vasopressin) in the BC-treated groups reduced by 31.9-40.0% ( p < 0.01) and 21.1-67.7% ( p < 0.01) compared to those in the constipation group, respectively. However, the secretion of excitability neurotransmitters (substance P and motilin) in the BC-treated groups was increased by 20.0-39.9% ( p < 0.01). The activities of ATPases in the colon of constipation rats were significantly weakened by BC administration ( p < 0.01). Histological morphology of the colon showed that BC supplementation could effectively increase the length of villus cells and the thickness of colonic mucosa and muscle ( p < 0.01). Moreover, BC supplementation could protect colonic smooth muscle cells against apoptosis. All of the findings suggest that BC supplementation effectively relieves constipation in rats and BC would be used as a great promising dietary fiber for alleviating constipation.


Subject(s)
Acetobacteraceae/metabolism , Cellulose/administration & dosage , Constipation/drug therapy , Diphenoxylate/adverse effects , Acetobacteraceae/chemistry , Animals , Aquaporins/metabolism , Cellulose/metabolism , Constipation/etiology , Constipation/metabolism , Constipation/physiopathology , Defecation/drug effects , Humans , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Male , Motilin/metabolism , Rats , Rats, Sprague-Dawley , Substance P/metabolism
19.
Acta Physiol (Oxf) ; 222(2)2018 02.
Article in English | MEDLINE | ID: mdl-28786555

ABSTRACT

AIM: Stomach contractions show two types of specific patterns in many species, that is migrating motor contraction (MMC) and postprandial contractions (PPCs), in the fasting and fed states respectively. We found gastric PPCs terminated with migrating strong contractions in humans, dogs and suncus. In this study, we reveal the detailed characteristics and physiological implications of these strong contractions of PPC. METHODS: Human, suncus and canine gastric contractions were recorded with a motility-monitoring ingestible capsule and a strain-gauge force transducer. The response of motilin and ghrelin and its receptor antagonist on the contractions were studied by using free-moving suncus. RESULTS: Strong gastric contractions were observed at the end of a PPC in human, dog and suncus models, and we tentatively designated this contraction to be a postprandial giant contraction (PPGC). In the suncus, the PPGC showed the same property as those of a phase III contraction of MMC (PIII-MMC) in the duration, motility index and response to motilin or ghrelin antagonist administration. Ghrelin antagonist administration in the latter half of the PPC (LH-PPC) attenuated gastric contraction prolonged the duration of occurrence of PPGC, as found in PII-MMC. CONCLUSION: It is thought that the first half of the PPC changed to PII-MMC and then terminated with PIII-MMC, suggesting that PPC consists of a digestive phase (the first half of the PPC) and a discharge phase (LH-PPC) and that LH-PPC is coincident with MMC. In this study, we propose a new approach for the understanding of postprandial contractions.


Subject(s)
Gastrointestinal Motility/physiology , Ghrelin/metabolism , Motilin/metabolism , Postprandial Period/physiology , Shrews/physiology , Animals , Dogs , Humans , Muscle Contraction/physiology , Stomach/physiology
20.
Am J Physiol Regul Integr Comp Physiol ; 313(2): R149-R157, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28566304

ABSTRACT

Patients with chronic kidney disease (CKD) commonly complain upper gastrointestinal (GI) symptoms, especially anorexia. Hemodialysis (HD) has been noted to improve GI symptoms; however, the underlying mechanisms are unclear. This study was designed 1) to study effects of HD on GI symptoms and gastric slow waves; and 2) to investigate possible roles of ghrelin and glucagon-like peptide-1 (GLP-1): the study recruited 13 healthy controls, 20 CKD patients without HD (CKD group), and 18 CKD patients with HD (HD group). Dyspeptic symptoms, autonomic functions, gastric slow waves, and plasma level of ghrelin and GLP-1 were analyzed. First, the CKD patients with HD showed markedly lower scores of anorexia (0.6 ± 0.2 vs. 3.2 ± 0.4, P < 0.001) compared with patients without HD. Second, the CKD group but not HD group showed a significant reduction (25.6%) in the percentage of normal gastric slow waves, compared with controls. Third, the CKD group exhibited a significantly lower ghrelin level compared with the HD group (26.8 ± 0.9 vs. 34.1 ± 2.3 ng/l, P < 0.02) and a higher GLP-1 level (29.4 ± 2.8 vs. 20.0 ± 2.1 pmol/l, P < 0.05) compared with controls. Moreover, the percentage of normal slow waves was positively correlated with ghrelin (r = 0.385, P = 0.019) but negatively correlated with GLP-1 (r = -0.558, P < 0.001) in all CKD patients. Hemodialysis improves upper GI symptoms and gastric slow waves in CKD patients. An increase in ghrelin and a decrease in GLP-1 might be involved in the HD-induced improvement in gastric slow waves.


Subject(s)
Gastrointestinal Hormones/metabolism , Motilin/metabolism , Myoelectric Complex, Migrating , Renal Dialysis , Renal Insufficiency, Chronic/physiopathology , Renal Insufficiency, Chronic/therapy , Adult , Aged , Female , Gastrointestinal Transit , Ghrelin/metabolism , Glucagon-Like Peptide 1/metabolism , Humans , Male , Middle Aged , Retrospective Studies , Stomach/physiopathology , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL