Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Neurogastroenterol Motil ; 36(5): e14764, 2024 May.
Article in English | MEDLINE | ID: mdl-38361111

ABSTRACT

BACKGROUND: Accelerated gastric emptying (GE) is a trait seen in obesity. Mutations in the hypothalamic leptin-melanocortin 4 receptor (Leptin-MC4R) pathway have been associated with obesity. We sought to investigate the association of leptin-MC4R pathway variants and GE in patients with obesity. METHODS: This is a cross-sectional study of patients with a history of severe obesity that were genotyped and completed a GE test by scintigraphy. We evaluated the percentage of GE (GE %) at 2 and 4 h between both groups using ANCOVA with weight and sex as covariates. We subdivide patients into carriers based on the location of the identified variants (i.e., upstream or downstream of the Leptin-MC4R pathway) and compared them with noncarriers using ANOVA. Results are presented as mean and standard deviation (± SD). KEY RESULTS: A total of 95 patients; nine carriers (67% females; 39.78 ± 12.33 years; BMI: 49.14 ± 12.96 kg/m2) and 86 noncarriers (87% female; 49.98 ± 13.74 years; BMI: 40.75 ± 6.29 kg/m2) were included. At 2 and 4 h, carriers had a delayed GE when compared noncarriers (p = 0.03 and p = 0.005, respectively). In carriers, when compared upstream carriers vs. downstream carriers vs. noncarriers by location there was a significant difference in GE among groups at 2 h and at 4 h (p = 0.02 and p = 0.01, respectively). CONCLUSIONS & INFERENCES: Carriers of heterozygous variants in the Leptin-MC4R pathway had a delayed GE compared to noncarriers. These findings point the important relationship between the Leptin-MC4R pathway and gastric motility.


Subject(s)
Gastric Emptying , Leptin , Obesity , Receptor, Melanocortin, Type 4 , Humans , Leptin/genetics , Female , Male , Gastric Emptying/physiology , Gastric Emptying/genetics , Adult , Cross-Sectional Studies , Middle Aged , Receptor, Melanocortin, Type 4/genetics , Obesity/genetics , Obesity/physiopathology , Signal Transduction
2.
Sci Rep ; 11(1): 22462, 2021 11 17.
Article in English | MEDLINE | ID: mdl-34789790

ABSTRACT

We examined the relationship between zonulin and gastric motility in critical care patients and a translational mouse model of systemic inflammation. Gastric motility and haptoglobin (HP) 2 isoform quantification, proxy for zonulin, were examined in patients. Inflammation was triggered by lipopolysaccharide (LPS) injection in C57Bl/6 zonulin transgenic mouse (Ztm) and wildtype (WT) mice as controls, and gastro-duodenal transit was examined by fluorescein-isothiocyanate, 6 and 12 h after LPS-injection. Serum cytokines and zonulin protein levels, and zonulin gastric-duodenal mRNA expression were examined. Eight of 20 patients [14 years, IQR (12.25, 18)] developed gastric dysmotility and were HP2 isoform-producing. HP2 correlated with gastric dysmotility (r = - 0.51, CI - 0.81 to 0.003, p = 0.048). LPS injection induced a time-dependent increase in IL-6 and KC-Gro levels in all mice (p < 0.0001). Gastric dysmotility was reduced similarly in Ztm and WT mice in a time-dependent manner. Ztm had 16% faster duodenal motility than WT mice 6H post-LPS, p = 0.01. Zonulin mRNA expression by delta cycle threshold (dCT) was higher in the stomach (9.7, SD 1.4) than the duodenum (13.9, SD 1.4) 6H post-LPS, p = 0.04. Serum zonulin protein levels were higher in LPS-injected mice compared to vehicle-injected animals in a time-dependent manner. Zonulin correlated with gastric dysmotility in patients. A mouse model had time-dependent gastro-duodenal dysmotility after LPS-injection that paralleled zonulin mRNA expression and protein levels.


Subject(s)
Gastric Emptying/genetics , Gastrointestinal Transit/genetics , Haptoglobins/metabolism , Protein Precursors/blood , Sepsis/blood , Adolescent , Animals , Child , Cohort Studies , Cytokines/blood , Disease Models, Animal , Female , Gastric Mucosa/metabolism , Haptoglobins/genetics , Humans , Intestinal Mucosa/metabolism , Lipopolysaccharides/administration & dosage , Lipopolysaccharides/adverse effects , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Precursors/genetics , Sepsis/chemically induced
3.
Eur J Endocrinol ; 185(4): R93-R101, 2021 Sep 01.
Article in English | MEDLINE | ID: mdl-34370694

ABSTRACT

In 2008, the first evidence of a new hormone called neuronostatin was published. The hormone was discovered using a bioinformatic method and found to originate from the same preprohormone as somatostatin. This small peptide hormone of 13 amino acids and a C-terminal amidation was soon found to exert pleiotropic physiological effects. In animal studies, neuronostatin has been shown to reduce food intake and delay gastric emptying and gastrointestinal transit. Furthermore, neuronostatin has been shown to affect glucose metabolism by increasing glucagon secretion during situations when glucose concentrations are low. Additionally, neuronostatin has been shown to affect neural tissue and cardiomyocytes by suppressing cardiac contractility. The effects of neuronostatin have not yet been delineated in humans, but if the effects found in animal studies translate to humans it could position neuronostatin as a promising target in the treatment of obesity, hypertension and diabetes. In this review, we describe the discovery of neuronostatin and the current understanding of its physiological role and potential therapeutic applicability.


Subject(s)
Peptide Hormones/physiology , Animals , Appetite Regulation/drug effects , Appetite Regulation/genetics , Diabetes Mellitus/genetics , Diabetes Mellitus/therapy , Gastric Emptying/drug effects , Gastric Emptying/genetics , Humans , Hypertension/genetics , Hypertension/therapy , Muscle Contraction/drug effects , Muscle Contraction/genetics , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Neurons/drug effects , Neurons/physiology , Obesity/genetics , Obesity/therapy , Peptide Hormones/pharmacology , Signal Transduction/drug effects , Somatostatin/chemistry , Somatostatin/pharmacology , Somatostatin/physiology
4.
Gastroenterology ; 160(7): 2451-2466.e19, 2021 06.
Article in English | MEDLINE | ID: mdl-33662386

ABSTRACT

BACKGROUND & AIMS: Gastrointestinal (GI) motility is regulated by serotonin (5-hydroxytryptamine [5-HT]), which is primarily produced by enterochromaffin (EC) cells in the GI tract. However, the precise roles of EC cell-derived 5-HT in regulating gastric motility remain a major point of conjecture. Using a novel transgenic mouse line, we investigated the distribution of EC cells and the pathophysiologic roles of 5-HT deficiency in gastric motility in mice and humans. METHODS: We developed an inducible, EC cell-specific Tph1CreERT2/+ mouse, which was used to generate a reporter mouse line, Tph1-tdTom, and an EC cell-depleted line, Tph1-DTA. We examined EC cell distribution, morphology, and subpopulations in reporter mice. GI motility was measured in vivo and ex vivo in EC cell-depleted mice. Additionally, we evaluated 5-HT content in biopsy and plasma specimens from patients with idiopathic gastroparesis (IG). RESULTS: Tph1-tdTom mice showed EC cells that were heterogeneously distributed throughout the GI tract with the greatest abundance in the antrum and proximal colon. Two subpopulations of EC cells were identified in the gut: self-renewal cells located at the base of the crypt and mature cells observed in the villi. Tph1-DTA mice displayed delayed gastric emptying, total GI transit, and colonic transit. These gut motility alterations were reversed by exogenous provision of 5-HT. Patients with IG had a significant reduction of antral EC cell numbers and 5-HT content, which negatively correlated with gastric emptying rate. CONCLUSIONS: The Tph1CreERT2/+ mouse provides a powerful tool to study the functional roles of EC cells in the GI tract. Our findings suggest a new pathophysiologic mechanism of 5-HT deficiency in IG.


Subject(s)
Gastric Emptying/genetics , Gastrointestinal Transit/genetics , Serotonin/deficiency , Animals , Cell Line , Enterochromaffin Cells/physiology , Humans , Mice , Mice, Transgenic , Tryptophan Hydroxylase/metabolism
5.
JCI Insight ; 6(2)2021 01 25.
Article in English | MEDLINE | ID: mdl-33491664

ABSTRACT

Hindered by a limited understanding of the mechanisms responsible for diabetic gastroenteropathy (DGE), management is symptomatic. We investigated the duodenal mucosal expression of protein-coding genes and microRNAs (miRNA) in DGE and related them to clinical features. The diabetic phenotype, gastric emptying, mRNA, and miRNA expression and ultrastructure of duodenal mucosal biopsies were compared in 39 DGE patients and 21 controls. Among 3175 differentially expressed genes (FDR < 0.05), several mitochondrial DNA-encoded (mtDNA-encoded) genes (12 of 13 protein coding genes involved in oxidative phosphorylation [OXPHOS], both rRNAs and 9 of 22 transfer RNAs) were downregulated; conversely, nuclear DNA-encoded (nDNA-encoded) mitochondrial genes (OXPHOS) were upregulated in DGE. The promoters of differentially expressed genes were enriched in motifs for transcription factors (e.g., NRF1), which regulate mitochondrial biogenesis. Seventeen of 30 differentially expressed miRNAs targeted differentially expressed mitochondrial genes. Mitochondrial density was reduced and correlated with expression of 9 mtDNA OXPHOS genes. Uncovered by principal component (PC) analysis of 70 OXPHOS genes, PC1 was associated with neuropathy (P = 0.01) and delayed gastric emptying (P < 0.05). In DGE, mtDNA- and nDNA-encoded mitochondrial genes are reduced and increased - associated with reduced mitochondrial density, neuropathy, and delayed gastric emptying - and correlated with cognate miRNAs. These findings suggest that mitochondrial disturbances may contribute to delayed gastric emptying in DGE.


Subject(s)
Diabetes Complications/etiology , Diabetes Complications/genetics , Gastroparesis/etiology , Gastroparesis/genetics , Genes, Mitochondrial , Adult , Case-Control Studies , DNA, Mitochondrial/genetics , Diabetes Complications/physiopathology , Duodenum/physiopathology , Duodenum/ultrastructure , Female , Gastric Emptying/genetics , Gastric Emptying/physiology , Gene Expression , Humans , Intestinal Mucosa/physiopathology , Intestinal Mucosa/ultrastructure , Male , MicroRNAs/genetics , Microscopy, Electron, Transmission , Middle Aged , Mitochondria/ultrastructure , Oxidative Phosphorylation , Promoter Regions, Genetic , RNA, Messenger/genetics
6.
Clin Transl Gastroenterol ; 11(3): e00136, 2020 03.
Article in English | MEDLINE | ID: mdl-32352713

ABSTRACT

INTRODUCTION: Epigenetic modifications have been implicated to mediate several complications of diabetes mellitus (DM), especially nephropathy and retinopathy. Our aim was to ascertain whether epigenetic alterations in whole blood discriminate among patients with DM with normal, delayed, and rapid gastric emptying (GE). METHODS: Using the ChIP-seq (chromatin immunoprecipitation combined with next-generation sequencing) assays, we compared the genome-wide enrichment of 3 histone modifications (i.e., H3K4me3, H3K9ac, and H3K27ac) in buffy coats from 20 diabetic patients with gastrointestinal symptoms and normal (n = 6), delayed (n = 8), or rapid (n = 6) GE. RESULTS: Between patients with DM with delayed vs normal GE, there were 108 and 54 genes that were differentially bound (false discovery rate < 0.05) with H3K27ac and H3K9ac, respectively; 100 genes were differentially bound with H3K9ac in patients with rapid vs normal GE. The differentially bound genes with H3K27ac were functionally linked to the type 2 immune response, particularly Th2 cell activation and function (e.g., CCR3, CRLF2, CXCR4, IL5RA, and IL1RL1) and glucose homeostasis (FBP-1, PDE4A, and CMKLR1). For H3K9ac, the differentially occupied genes were related to T-cell development and function (e.g., ICOS and CCR3) and innate immunity (RELB, CD300LB, and CLEC2D). Compared with normal GE, rapid GE had differential H3K9ac peaks at the promoter site of diverse immunity-related genes (e.g., TNFRSF25 and CXCR4) and genes related to insulin resistance and glucose metabolism. Motif analysis disclosed enrichment of binding sites for transcription factors relevant to the pathogenesis and complications of DM. DISCUSSION: GE disturbances in DM are associated with epigenetic alterations that pertain to dysimmunity, glucose metabolism, and other complications of DM.


Subject(s)
Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 2/complications , Epigenesis, Genetic , Gastric Emptying/genetics , Gastrointestinal Diseases/genetics , Adult , Blood Buffy Coat , Chromatin Immunoprecipitation Sequencing , Computational Biology , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/genetics , Female , Gastrointestinal Diseases/blood , Gastrointestinal Diseases/diagnosis , Glucose/metabolism , Histone Code/genetics , Histones/genetics , Humans , Insulin Resistance/genetics , Male , Middle Aged , Promoter Regions, Genetic/genetics , Severity of Illness Index
7.
Neurogastroenterol Motil ; 32(1): e13724, 2020 01.
Article in English | MEDLINE | ID: mdl-31691451

ABSTRACT

BACKGROUND: Nutrient-mediated release of cholecystokinin and glucagon-like peptide-1 (GLP-1) regulates gastric emptying (GE) via duodenogastric feedback mechanisms; GLP-1 also regulates postprandial insulin secretion. Some patients with functional upper gastrointestinal symptoms have impaired glucose tolerance during enteral dextrose infusion. Our hypothesis was that variants in CCK, GLP-1, and TCF7L2 (transcription factor 7-like 2 locus), which is associated with greatest genetic risk for development of type 2 diabetes mellitus, are associated with GE and independently with glucose tolerance. Our aims were to evaluate the associations between these GE, glucose tolerance, and these single nucleotide polymorphisms (SNPs). METHODS: Genetic variants, scintigraphic GE of solids, plasma glucose, insulin, and GLP-1 during enteral dextrose infusion (75gm over 2 hours) were measured. GE and enteral dextrose infusion were, respectively, evaluated in 44 (27 controls and 17 patients with functional dyspepsia or nausea) and 42 (28 controls, 14 patients) participants; of these, 51 participants consented to assessment of SNPs. Four functional SNPs were studied: rs6923761 and rs1042044 at GLP-1 receptor, rs7903146 (TCF7L2), and rs1800857 (CCK receptor). KEY RESULTS: Gastric emptying was normal in 38, rapid in 4, and delayed in two participants; 38 had normal, and four had impaired glucose tolerance. The T allele at rs7903146 (TCF7L2) was non-significantly associated (P = .14) with faster GE. The associations between SNPs and demographic variables, GE thalf , glucose tolerance and plasma GLP1 levels were not significant. CONCLUSIONS & INFERENCES: There is a trend toward an association between faster GE and the diabetes-associated allele at rs7903146 in TCF7L2. However, these SNPs were not associated with plasma glucose or GLP1 concentrations during enteral dextrose infusion.


Subject(s)
Gastric Emptying/genetics , Glucagon-Like Peptide 1/genetics , Glucose Intolerance/genetics , Receptors, Cholecystokinin/genetics , Transcription Factor 7-Like 2 Protein/genetics , Adult , Female , Gastrointestinal Diseases/genetics , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide
8.
Br Poult Sci ; 60(6): 649-658, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31469320

ABSTRACT

1. A previous whole-genome association analysis has identified the motilin receptor gene (MLNR), which regulates gastrointestinal motility and gastric emptying, as a candidate gene related to chicken growth.2. MLNR mRNA was expressed in all tissues tested, and the expression level in digestive tissues was greater than in other tissues. Expression levels in the pancreas, duodenum and glandular stomach at day old and one, two and three weeks of age indicated a possible correlation with the digestive system. This suggested that the MLNR gene plays a central role in gastrointestinal tract function and affects the growth and development of chickens. Moreover, there was a significant difference in expression in the glandular stomach tissue between Ross 308 and Gushi chickens at six weeks of age.3. Re-sequencing revealed an 86-bp insertion/deletion polymorphism in the downstream region of the MLNR gene. The mutation locus was genotyped in 2,261 individuals from nine different chicken breeds. MLNR expression levels in the glandular stomach of chickens with DD genotypes were greater than those in chickens with the ID and II genotypes. The DD genotype was the most dominant genotype in commercial broiler's (Ross 308 and Arbor Acres broilers), and the D allele frequency in these breeds exceeded 91%. The deletion mutation tended towards fixation in commercial broilers.4. Association with growth and carcass traits analysed in a Gushi-Anka F2 intercrossed population, showed that the DD genotype was significantly associated with the greatest growth and carcass trait values, whereas values associated with the II genotype were the lowest in the F2 reciprocal cross chickens.5. The results suggest that the mutation is strongly associated with growth related traits and it is likely to be useful for marker-assisted selection of chickens.


Subject(s)
Chickens/genetics , INDEL Mutation , Receptors, Gastrointestinal Hormone/genetics , Receptors, Neuropeptide/genetics , Animals , Chickens/anatomy & histology , Chickens/growth & development , Crosses, Genetic , DNA, Complementary/blood , DNA, Complementary/isolation & purification , Duodenum/metabolism , Female , Gastric Emptying/genetics , Gastric Mucosa/metabolism , Gastrointestinal Motility/genetics , INDEL Mutation/genetics , Male , Pancreas/metabolism , RNA, Messenger/analysis , RNA, Messenger/biosynthesis
9.
Peptides ; 121: 170124, 2019 11.
Article in English | MEDLINE | ID: mdl-31415798

ABSTRACT

Urocortin3 (UCN3), the newest member of corticotrophin releasing hormone (CRH) family polypeptides, is an anorexic factor discovered in 2001, which has a strong inhibitory effect on animal appetite regulation. UCN3 is widely distributed in various tissues of animals and has many biological functions. Based on the research progress of UCN3 on mammals and non-mammals, this paper summarized the discovery, tissue distribution, appetite regulation and mechanism of UCN3 in animals, in order to provide a reference for feeding regulation and growth in mammals and fish in further research and production.


Subject(s)
Adrenocorticotropic Hormone/genetics , Appetite Regulation/genetics , Appetite/genetics , Gastric Emptying/genetics , Urocortins/genetics , Adrenocorticotropic Hormone/metabolism , Animals , Chickens , Cholecystokinin/genetics , Cholecystokinin/metabolism , Gene Expression Regulation , Ghrelin/genetics , Ghrelin/metabolism , Humans , Mice , Neuropeptide Y/genetics , Neuropeptide Y/metabolism , Pro-Opiomelanocortin/genetics , Pro-Opiomelanocortin/metabolism , Rats , Receptors, Corticotropin-Releasing Hormone/genetics , Receptors, Corticotropin-Releasing Hormone/metabolism , Signal Transduction/genetics , Urocortins/metabolism , Xenopus laevis , Zebrafish
10.
Free Radic Biol Med ; 135: 132-143, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30831189

ABSTRACT

Diabetic gastroparesis (GP) is a clinical syndrome characterized by delayed gastric emptying (DGE). Loss of Nrf2 (Nuclear factor (erythroid-derived 2)-like 2) led to reduced nNOSα mediated gastric motility and DGE. The molecular signaling of cinnamaldehyde (CNM) mediated Nrf2 activation and its mechanistic role on DGE were further investigated in obese/T2D female mice. Adult female homozygous Nfe2l2-/- (C57BL/6J) and their wild-type (WT) littermates (Nfe2l2+/+) mice were fed with high fat diet (HFD; Obese/T2D model), or normal diet (ND) with or without CNM (50 mg/kg b.w; i.p). Supplementation of CNM attenuated (p < 0.05) DGE in WT female but not in Nrf2 KO Obese/T2D mice. CNM (1) normalized serum estradiol-17ß levels, (2) induced gastric Nrf2 and phase II antioxidant enzymes through extracellular signal-regulated kinase, (ERK)/c-Jun N-terminal kinase (JNK)/p38 mitogen-activated protein kinase (MAPK), (3) reduced glucose synthase kinase 3 beta (GSK3ß) and aryl hydrocarbon receptor (AhR) and this was associated with (4) increased estrogen receptor expression, BH4 (Cofactor of nNOS) biosynthesis enzyme GCH-1 and nNOSα dimerization in WT Obese/T2 diabetic female mice. In addition, CNM restored impaired nitrergic relaxation in hyperglycemic conditions. These findings emphasize the importance of Nrf2 in maintaining nNOSα mediated GE and may have a translational relevance to treat obese/diabetic gastroparesis in women.


Subject(s)
Acrolein/analogs & derivatives , Diabetes Complications/genetics , Gastroparesis/genetics , NF-E2-Related Factor 2/genetics , Obesity/genetics , Acrolein/pharmacology , Animals , Antioxidants/metabolism , Antioxidants/pharmacology , Diabetes Complications/drug therapy , Diabetes Complications/metabolism , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Diet, High-Fat/adverse effects , Gastric Emptying/drug effects , Gastric Emptying/genetics , Gastroparesis/drug therapy , Gastroparesis/etiology , Gastroparesis/metabolism , Humans , MAP Kinase Signaling System/drug effects , Mice , Muscle Relaxation/drug effects , Muscle Relaxation/genetics , NF-E2-Related Factor 2/metabolism , Nitric Oxide Synthase Type I/genetics , Obesity/complications , Obesity/drug therapy , Obesity/metabolism , Stomach/pathology , p38 Mitogen-Activated Protein Kinases/genetics
11.
Neurogastroenterol Motil ; 30(7): e13313, 2018 07.
Article in English | MEDLINE | ID: mdl-29488276

ABSTRACT

BACKGROUND: Weight loss in response to the long-acting GLP-1 receptor (GLP1R) analog, liraglutide, is correlated with delay in gastric-emptying (GE). The aim of this pilot study was to assess whether specific genetic variants in GLP1R or TCF7L2 are associated with delayed GE and weight loss in obese patients treated with liraglutide or the short-acting GLP-1 agonist, exenatide. METHODS: We evaluated in obese individuals the associations of genetic variations of GLP1R (rs6923761) and TCF7L2 (rs7903146) on GE T1/2 and weight from two trials that evaluated separately exenatide, 5 µg BID for 30 days, or liraglutide, 3 mg daily for 5 weeks. Data were analyzed using the dominant genetic model and intention-to-treat analysis. KEY RESULTS: There was a significant correlation between changes in weight and GE T1/2 (rs  = -.382, P = .004). GLP1R rs6923761 minor allele A (AA_AG) carriers who received either exenatide or liraglutide had greater delay in GE T1/2 relative to baseline (117.9 ± 27.5 [SEM] minutes and 128.9 ± 38.32 minutes) compared to GG genotype (95.8 ± 30.4 minutes and 61.4 ± 21.4 minutes, respectively; P = .11). There was a non-significant difference in weight loss based on GLP1R rs6923761 genotype after 5 weeks of treatment. There were no significant correlations with TCF7L2 (rs7903146) genotype. CONCLUSIONS & INFERENCES: The minor A allele of GLP1R (rs6923761) is associated with greater delay in GE T1/2 in response to liraglutide and exenatide. These studies provide data to plan pharmacogenetics testing of the hypothesis that GLP1R (rs6923761) influences weight loss in response to GLP1R agonists.


Subject(s)
Exenatide/pharmacology , Gastric Emptying/genetics , Genetic Variation/genetics , Glucagon-Like Peptide-1 Receptor/genetics , Liraglutide/pharmacology , Pharmacogenetics/methods , Adult , Alleles , Double-Blind Method , Exenatide/therapeutic use , Female , Gastric Emptying/drug effects , Genetic Variation/drug effects , Glucagon-Like Peptide-1 Receptor/agonists , Humans , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Liraglutide/therapeutic use , Male , Middle Aged , Obesity/drug therapy , Obesity/genetics , Pilot Projects , Weight Loss/drug effects , Weight Loss/physiology
12.
PLoS One ; 12(11): e0187772, 2017.
Article in English | MEDLINE | ID: mdl-29161307

ABSTRACT

BACKGROUND: Idiopathic and diabetic gastroparesis in Homo sapiens cause significant morbidity. Etiology or risk factors have not been clearly identified. Failure to sustain elevated heme oxygenase-1 (HO1) expression is associated with delayed gastric emptying in diabetic mice and polymorphisms in the HO1 gene (HMOX1, NCBI Gene ID:3162) are associated with worse outcomes in other diseases. AIM: Our hypothesis was that longer polyGT alleles are more common in the HMOX1 genes of individuals with gastroparesis than in controls without upper gastrointestinal motility disorders. METHODS: Repeat length was determined in genomic DNA. Controls with diabetes (84 type 1, 84 type 2) and without diabetes (n = 170) were compared to diabetic gastroparetics (99 type 1, 72 type 2) and idiopathic gastroparetics (n = 234). Correlations of repeat lengths with clinical symptom sub-scores on the gastroparesis cardinal symptom index (GCSI) were done. Statistical analyses of short (<29), medium and long (>32) repeat alleles and differences in allele length were used to test for associations with gastroparesis. RESULTS: The distribution of allele lengths was different between groups (P = 0.016). Allele lengths were longest in type 2 diabetics with gastroparesis (29.18±0.35, mean ± SEM) and longer in gastroparetics compared to non-diabetic controls (28.50±0.14 vs 27.64±0.20 GT repeats/allele, P = 0.0008). Type 2 diabetic controls had longer alleles than non-diabetic controls. In all gastroparetic groups, allele lengths were longer in African Americans compared to other racial groups, differences in the proportion of African Americans in the groups accounted for the differences between gastroparetics and controls. Diabetic gastroparetics with 1 or 2 long alleles had worse GCSI nausea sub-scores (3.30±0.23) as compared to those with 0 long alleles (2.66±0.12), P = 0.022. CONCLUSIONS: Longer poly-GT repeats in the HMOX1 gene are more common in African Americans with gastroparesis. Nausea symptoms are worse in subjects with longer alleles.


Subject(s)
Diabetes Mellitus/genetics , Gastroparesis/genetics , Heme Oxygenase-1/genetics , Tandem Repeat Sequences/genetics , Adult , Black or African American/genetics , Aged , Alleles , Animals , Diabetes Complications/genetics , Diabetes Complications/pathology , Diabetes Mellitus/pathology , Female , Gastric Emptying/genetics , Gastroparesis/pathology , Humans , Male , Mice , Middle Aged , Polymorphism, Genetic
13.
J Clin Gastroenterol ; 51(2): 136-144, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27092429

ABSTRACT

GOALS: The goals of the study were to investigate in both postprandial distress syndrome (PDS) and epigastric pain syndrome (EPS) the gastric electrical activity and the gastric emptying (GE) time together with the circulating concentrations of motilin, somatostatin, corticotrophin-releasing factor, and neurotensin, and to establish whether the genetic variability in the neurotensin system genes differs between these 2 categories of functional dyspepsia (FD). BACKGROUND: The current FD classification is based on symptoms and it has been proven not to be completely satisfying because of a high degree of symptom overlap between subgroups. STUDY: Gastric electrical activity was evaluated by cutaneous electrogastrography: the GE time by C-octanoic acid breast test. Circulating concentrations of gut peptides were measured by a radioimmunoassay. NTS 479 A/G and NTSR1 rs6090453 SNPs were evaluated by PCR and endonuclease digestion. RESULTS: Fifty-four FD patients (50 female/4 male) were studied. Using a symptom questionnaire, 42 patients were classified as PDS and 12 as EPS, although an overlap between the symptom profiles of the 2 subgroups was recorded. The electrogastrographic parameters (the postprandial instability coefficient of dominant frequency, the dominant power, and the power ratio) were significantly different between the subgroups, whereas the GE time did not differ significantly. In addition, EPS was characterized by a different gut peptide profile compared with PDS. Finally, neurotensin polymorphism was shown to be associated with neurotensin levels. This evidence deserves further studies in consideration of an analgesic role of neurotensin. CONCLUSIONS: Analysis of gut peptide profiles could represent an interesting tool to enhance FD diagnosis and overcome limitations due to a distinction based solely on symptoms.


Subject(s)
Abdominal Pain/diagnosis , Dyspepsia/diagnosis , Peptides/blood , Postprandial Period/physiology , Symptom Assessment/methods , Abdominal Pain/etiology , Abdominal Pain/physiopathology , Adrenocorticotropic Hormone/blood , Adrenocorticotropic Hormone/genetics , Adult , Aged , Caprylates/analysis , Diagnosis, Differential , Dyspepsia/complications , Dyspepsia/physiopathology , Electric Conductivity , Female , Gastric Emptying/genetics , Humans , Male , Middle Aged , Motilin/blood , Motilin/genetics , Neurotensin/blood , Neurotensin/genetics , Polymorphism, Genetic , Somatostatin/blood , Somatostatin/genetics , Stomach/physiopathology , Syndrome , Time Factors
14.
Diabetes ; 65(1): 34-43, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26470787

ABSTRACT

Nutrient stimulation of enteroendocrine L cells induces the release of the incretin and satiating peptide glucagon-like peptide 1 (GLP-1). The vagus nerve innervates visceral organs and may contribute to the mediation of gut-derived GLP-1's effects on food intake, energy homeostasis, and glycemic control. To test the hypothesis that vagal afferent neuron (VAN) GLP-1 receptors (GLP-1Rs) are necessary for these effects of endogenous GLP-1, we established a novel bilateral nodose ganglia injection technique to deliver a lentiviral vector and to knock down VAN GLP-1Rs in male Sprague Dawley rats. We found that a full expression of VAN GLP-1Rs is not necessary for the maintenance of long-term energy balance in normal eating conditions. VAN GLP-1R knockdown (kd) did, however, increase meal size and accelerated gastric emptying. Moreover, postmeal glycemia was elevated and insulin release was blunted in GLP-1R kd rats, suggesting that VAN GLP-1Rs are physiological contributors to the neuroincretin effect after a meal. Collectively, our results highlight a crucial role for the VANs in mediating the effects of endogenous GLP-1 on food intake and glycemia and may promote the further development of GLP-1-based therapies.


Subject(s)
Blood Glucose/metabolism , Eating/genetics , Energy Metabolism/genetics , Glucagon-Like Peptide-1 Receptor/genetics , Insulin/metabolism , Neurons, Afferent/metabolism , Nodose Ganglion/metabolism , Animals , Gastric Emptying/genetics , Gene Knockdown Techniques , Insulin Secretion , Intestines/innervation , Male , Postprandial Period , Rats , Rats, Sprague-Dawley , Vagus Nerve/metabolism
15.
Eur J Nutr ; 55(4): 1573-81, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26160548

ABSTRACT

PURPOSE: There are wide inter-individual differences in glycemic response (GR). We aimed to examine key digestive parameters that influence inter-individual and ethnic differences in GR in healthy Asian individuals. METHODS: Seventy-five healthy male subjects (25 Chinese, 25 Malays, and 25 Asian-Indians) were served equivalent available carbohydrate amounts (50 g) of jasmine rice (JR) and basmati rice (BR) on separate occasions. Postprandial blood glucose concentrations were measured at fasting (-5 and 0 min) and at 15- to 30-min interval over 180 min. Mastication parameters (number of chews per mouth and chewing time per mouthful), saliva α-amylase activity, AMY1 gene copy numbers and gastric emptying rate were measured to investigate their relationships with GR. RESULTS: The GR for jasmine rice was significantly higher than for basmati rice (P < 0.001). The median number of AMY1 gene copies was 6, with a range of 2-15. There was a significant positive relationship between AMY1 copy number and α-amylase activity (P = 0.002). There were no significant ethnic differences in GR. For both rice varieties, the number of chews per mouthful was positively associated with the GR (JR, P = 0.011; BR, P = 0.005), while chewing time per mouthful showed a negative association (JR, P = 0.039; BR, P = 0.016). Ethnicity, salivary α-amylase activity, particle size distribution, gastric emptying rate and AMY1 gene copy numbers were not significant contributors to GR (P > 0.05). CONCLUSION: Mastication parameters contribute significantly to GR. Eating slowly and having larger food boluses before swallowing (less chewing), both potentially modifiable, may be beneficial in glycemic control.


Subject(s)
Asian People , Blood Glucose/metabolism , Digestion/ethnology , Glycemic Index , Mastication , Adiposity , Adult , Blood Pressure , Body Mass Index , China , Cross-Over Studies , Dietary Carbohydrates/administration & dosage , Dietary Carbohydrates/analysis , Gastric Emptying/genetics , Gastric Emptying/physiology , Gene Dosage , Humans , India , Linear Models , Malaysia , Male , Oryza/chemistry , Oryza/classification , Particle Size , Postprandial Period , Salivary alpha-Amylases/genetics , Waist Circumference , Young Adult
16.
Neurogastroenterol Motil ; 28(1): 36-42, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26486522

ABSTRACT

BACKGROUND: Severe burn injury has been demonstrated to delay gastric emptying. The aim of this study was to investigate effects and cellular mechanisms of auricular electroacupuncture (AEA) at the acupoints innervated by the auricular branch of vagus nerve on burn-induced gastric dysmotility in rats. METHODS: Propranolol (ß-adrenoceptor antagonist) was injected intraperitoneally after the rats underwent burn injury. All experiments were performed 6 h following burn/sham burn injury. AEA was performed at bilateral auricular acupoints for 45 min. Electrocardiogram was recorded for 30 min. Plasma hormones were measured; cyclooxygenase (COX)-2 expressions in gastric tissue were measured using western blotting and real-time RT-PCR. KEY RESULTS: (i) Burn injury delayed gastric emptying (p = 0.006) and AEA increased gastric emptying by 49% (p = 0.045). (ii) Burn injury evoked a significant elevation in plasma noradrenaline, which was suppressed by AEA. (iii) Burn injury significantly increased protein and mRNA expressions of COX-2 in gastric fundus and antrum. AEA suppressed burn-induced increase in protein expressions, but not mRNA expressions of COX-2. CONCLUSIONS & INFERENCES: Burn injury delays gastric emptying by up-regulating COX-2 attributed to sympathetic overactivity. AEA improves burn-induced delay in gastric emptying, possibly mediated via the sympathetic-COX-2 pathway.


Subject(s)
Burns/therapy , Cyclooxygenase 2/genetics , Electroacupuncture/methods , Gastric Emptying/genetics , Gastric Mucosa/metabolism , Gastroparesis/therapy , RNA, Messenger/metabolism , Vagus Nerve Stimulation/methods , Acupuncture, Ear/methods , Adrenergic beta-Antagonists/pharmacology , Animals , Burns/complications , Burns/genetics , Cyclooxygenase 2/metabolism , Gastric Emptying/drug effects , Gastric Fundus/drug effects , Gastric Fundus/metabolism , Gastroparesis/etiology , Gastroparesis/genetics , Propranolol/pharmacology , Pyloric Antrum/drug effects , Pyloric Antrum/metabolism , RNA, Messenger/drug effects , Rats , Stomach/drug effects , Up-Regulation
17.
J Pain Palliat Care Pharmacother ; 29(2): 115-9, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26095480

ABSTRACT

Acetaminophen has been used as a tool for clinical and nonclinical experimental designs that evaluate gastric emptying because acetaminophen is not absorbed in stomach but efficiently absorbed from the small intestine. Published pharmacokinetic data of acetaminophen in subjects with normal gastric emptying vs. impaired gastric emptying (i.e., morphine treatment) were evaluated to select a key surrogate. Using Caverage (average concentration), computed from the exposure within the first hour, individual rank distribution was plotted across different studies. Caverage was highly correlated with Cmax (maximum concentration) in subjects with normal gastric emptying (R(2) = .7532) but not in those where gastric emptying was impaired (R(2) = .0213). The 50th percentile value of the distribution pattern of 1/Caverage in acetaminophen+morphine-treated group (coincided with the first shift in the slope) was considered as the cutoff point to figure out the impaired gastric emptying. The individual rank distribution plots for 1/Caverage across different studies supported similar trends in subjects with normal gastric emptying but showed a distinct distribution pattern in the cohort of impaired gastric emptying. Caverage, calculated within the first hour of dosing of acetaminophen (average concentration at 0-1 hour, C0-1havg), can be used as a key surrogate to distinguish the effects of gastric emptying on the absorption of acetaminophen. A 4 µg/mL C0-1havg of acetaminophen (dose: 1.5 g) may be used as cutoff point in future clinical investigations of acetaminophen to clarify the role of gastric emptying.


Subject(s)
Acetaminophen/pharmacokinetics , Analgesics, Non-Narcotic/pharmacokinetics , Gastric Emptying , Drug Interactions , Gastric Emptying/genetics , Humans , Intestinal Absorption , Morphine/pharmacology
18.
Obesity (Silver Spring) ; 23(4): 898-906, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25755013

ABSTRACT

OBJECTIVE: To examine the association of gene variants of uncoupling proteins (UCP)-2 and -3 with obesity and gastrointestinal (GI) traits. METHODS: In 255 overweight or obese adults, the associations of gene variants in UCP-2 (-3474, rs659366) and UCP-3 (rs1626521, rs2075577, rs15763) with body weight (BW) and GI traits were studied. Gene variants were genotyped by TaqMan® assay. The associations of genotypes with BW and GI traits (gastric emptying, gastric volume, satiety by buffet meal, satiation by nutrient drink test and GI hormones) were assessed using ANOVA corrected for false detection rate (FDR). RESULTS: A novel UCP-3 gene variant, rs1626521, was identified; it was associated with BW (P = 0.039), waist circumference (P = 0.035), and significantly higher postprandial gastric volume (P = 0.003) and calories ingested at buffet meal (P = 0.006, both significant with FDR). In a subgroup of 11 participants, rs1626521 was also associated with reduced mitochondrial bioenergetics efficiency in skeletal muscle (P = 0.051). In an in vitro study in HEK293 cells, rs1626521 reduced UCP-3 protein expression (P = 0.049). Associations detected between other genotypes and GI traits were nonsignificant with FDR. CONCLUSIONS: A newly identified functional variant (rs1626521) in UCP-3 affects postprandial gastric functions and satiety and may contribute to weight gain and alter human mitochondrial function.


Subject(s)
Gastric Emptying/genetics , Ion Channels/genetics , Mitochondrial Proteins/genetics , Obesity/genetics , Adult , Gastric Emptying/physiology , Genetic Association Studies , Genotype , Humans , Obesity/metabolism , Polymorphism, Single Nucleotide , Postprandial Period/physiology , Satiation/physiology , Uncoupling Protein 2 , Uncoupling Protein 3
19.
Sci Rep ; 5: 7919, 2015 Jan 21.
Article in English | MEDLINE | ID: mdl-25605129

ABSTRACT

Cinnamon extract is associated to different health benefits but the active ingredients or pathways are unknown. Cinnamaldehyde (CIN) imparts the characteristic flavor to cinnamon and is known to be the main agonist of transient receptor potential-ankyrin receptor 1 (TRPA1). Here, expression of TRPA1 in epithelial mouse stomach cells is described. After receiving a single-dose of CIN, mice significantly reduce cumulative food intake and gastric emptying rates. Co-localization of TRPA1 and ghrelin in enteroendocrine cells of the duodenum is observed both in vivo and in the MGN3-1 cell line, a ghrelin secreting cell model, where incubation with CIN up-regulates expression of TRPA1 and Insulin receptor genes. Ghrelin secreted in the culture medium was quantified following CIN stimulation and we observe that octanoyl and total ghrelin are significantly lower than in control conditions. Additionally, obese mice fed for five weeks with CIN-containing diet significantly reduce their cumulative body weight gain and improve glucose tolerance without detectable modification of insulin secretion. Finally, in adipose tissue up-regulation of genes related to fatty acid oxidation was observed. Taken together, the results confirm anti-hyperglycemic and anti-obesity effects of CIN opening a new approach to investigate how certain spice derived compounds regulate endogenous ghrelin release for therapeutic intervention.


Subject(s)
Acrolein/analogs & derivatives , Anti-Obesity Agents/pharmacology , Eating/drug effects , Gastric Emptying/drug effects , Ghrelin/metabolism , Hypoglycemic Agents/pharmacology , Acrolein/pharmacology , Animals , Cell Line , Eating/genetics , Epithelial Cells/metabolism , Gastric Emptying/genetics , Gastric Mucosa/metabolism , Gene Expression Regulation/drug effects , Ghrelin/genetics , Mice , Mice, Knockout , Mice, Obese , TRPA1 Cation Channel , Transient Receptor Potential Channels/biosynthesis , Transient Receptor Potential Channels/genetics
20.
Am J Physiol Gastrointest Liver Physiol ; 307(11): G1044-56, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25277798

ABSTRACT

The single-minded 2 (SIM2) protein is a basic helix-loop-helix transcription factor regulating central nervous system (CNS) development in Drosophila. In humans, SIM2 is located within the Down syndrome critical region on chromosome 21 and may be involved in the development of mental retardation phenotype in Down syndrome. In this study, knockout of SIM2 expression in mice resulted in a gas distention phenotype in the gastrointestinal tract. We found that SIM2 is required for the expression of all cryptdins and numerous other antimicrobial peptides (AMPs) expressed in the small intestine. The mechanism underlying how SIM2 controls AMP expression involves both direct and indirect regulations. For the cryptdin genes, SIM2 regulates their expression by modulating transcription factor 7-like 2, a crucial regulator in the Wnt/ß-catenin signaling pathway, while for other AMP genes, such as RegIIIγ, SIM2 directly activates their promoter activity. Our results establish that SIM2 is a crucial regulator in controlling expression of intestinal AMPs to maintain intestinal innate immunity against microbes.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Immunity, Innate/genetics , Intestine, Small/immunology , Alkaline Phosphatase/metabolism , Animals , Antimicrobial Cationic Peptides/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Count , Female , Gastric Emptying/genetics , Gastric Emptying/physiology , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/pathology , In Vitro Techniques , Intestinal Absorption/genetics , Intestinal Absorption/physiology , Intestine, Small/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Peristalsis/genetics , Peristalsis/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...