Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
3.
JCI Insight ; 7(19)2022 10 10.
Article in English | MEDLINE | ID: mdl-36099053

ABSTRACT

A high-fat diet (HFD) contributes to the increased incidence of colorectal cancer, but the mechanisms are unclear. We found that R-spondin 3 (Rspo3), a ligand for leucine-rich, repeat-containing GPCR 4 and 5 (LGR4 and LGR5), was the main subtype of R-spondins and was produced by myofibroblasts beneath the crypts in the intestine. HFD upregulated colonic Rspo3, LGR4, LGR5, and ß-catenin gene expression in specific pathogen-free rodents, but not in germ-free mice, and the upregulations were prevented by the bile acid (BA) binder cholestyramine or antibiotic treatment, indicating mediation by both BA and gut microbiota. Cholestyramine or antibiotic treatments prevented HFD-induced enrichment of members of the Lachnospiraceae and Rumincoccaceae, which can transform primary BA into secondary BA. Oral administration of deoxycholic acid (DCA), or inoculation of a combination of the BA deconjugator Lactobacillus plantarum and 7α-dehydroxylase-containing Clostridium scindens with an HFD to germ-free mice increased serum DCA and colonic Rspo3 mRNA levels, indicating that formation of secondary BA by gut microbiota is responsible for HFD-induced upregulation of Rspo3. In primary myofibroblasts, DCA increased Rspo3 mRNA via TGR5. Finally, we showed that cholestyramine or conditional deletion of Rspo3 prevented HFD- or DCA-induced intestinal proliferation. We conclude that secondary BA is responsible for HFD-induced upregulation of Rspo3, which, in turn, mediates HFD-induced intestinal epithelial proliferation.


Subject(s)
Bile Acids and Salts , Diet, High-Fat , Animals , Anti-Bacterial Agents , Cell Proliferation , Cholestyramine Resin , Deoxycholic Acid , Diet, High-Fat/adverse effects , Intestines , Leucine , Ligands , Mice , RNA, Messenger , Up-Regulation , beta Catenin/metabolism
4.
Gastroenterology ; 2022 08 04.
Article in English | MEDLINE | ID: mdl-35934059

ABSTRACT

This article has been withdrawn at the request of the author(s) and/or editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at https://www.elsevier.com/about/policies/article-withdrawal

5.
Zhonghua Nan Ke Xue ; 27(10): 927-933, 2021 10 20.
Article in Chinese | MEDLINE | ID: mdl-34914272

ABSTRACT

Prostate cancer (PCa) is a maligmancy with high morbidity and mortality. Bone metastasis is the main cause of short survival time and difficulties in the treatment and prevention of PCa. Previous findings of our team showed 155 bone-specific genes highly expressed in bone metastatic PC3 cells, which is considered to be the key to their adaptation to the bone micro-environment, proliferation and formation of metastatic tumor, and extensively exists in cancer metastasis in multiple systems. This review summarizes the published literature on the highly expressed bone-specific genes, focusing on the roles and values of these genes in the metastasis, progression, clinical diagnosis, treatment and prognosis of PCa, offering a prospect of the direction and targets in the studies of PCa bone metastasis so as to enrich the bone metastatic theories and clinical treatment principles of this disease in the future.


Subject(s)
Prostatic Neoplasms , Humans , Male , PC-3 Cells , Prostatic Neoplasms/genetics , Tumor Microenvironment
6.
JCI Insight ; 6(22)2021 11 22.
Article in English | MEDLINE | ID: mdl-34618688

ABSTRACT

Fermentable oligosaccharides, disaccharides, monosaccharides, and polyols (FODMAPs) are carbohydrates thought to contribute to the symptoms of IBS. A diet in high in FODMAPs (HFM) induces gastrointestinal symptoms in patients with irritable bowel syndrome (IBS), and a diet low in FODMAPs (LFM) improves symptoms in up to 60% of patients with IBS. However, the mechanism by which FODMAPs affect IBS symptoms is unclear. We showed that mice fed on a HFM diet have mast cell activation and colonic barrier loss. Using mast cell-deficient mice with and without mast cell reconstitution, we showed that HFM-mediated colonic barrier loss is dependent on TLR4-dependent mast cell activation. In in vitro studies, we demonstrated that IBS fecal supernatant stimulates mast cells significantly more compared with fecal supernatant from healthy controls. This effect of IBS fecal supernatant on mast cell stimulation is ameliorated in the absence of the TLR4 receptor and after a LFM diet. We found that a LFM diet improves colonic barrier function and reduces mast cell activation while decreasing fecal LPS levels. Our findings indicate that a HFM diet causes mast cell activation via LPS, which in turn leads to colonic barrier loss, and a LFM diet reverses these pathophysiologic mucosal changes.


Subject(s)
Fermentation/physiology , Gastrointestinal Diseases/diet therapy , Irritable Bowel Syndrome/diet therapy , Lipopolysaccharides/metabolism , Mast Cells/metabolism , Adult , Animals , Female , Humans , Male , Mice , Treatment Outcome
7.
Int J Biol Macromol ; 168: 395-402, 2021 Jan 31.
Article in English | MEDLINE | ID: mdl-33275979

ABSTRACT

Spider silk, which is composed of diverse silk proteins (spidroin), is a kind of natural high-mass biomaterial with great potential. However, due to the complexity of both the structure and the composition of the spidroins in natural spider silk, application of this valuable biomass is still limited to date. There are diverse kinds of spider silk in the orb-weaving spider with different mechanical and structural characteristics. In order to systematically illustrate the landscape of all the different spidrons, here we chose Araneus ventricosus, an orb-weaving spider with superior silk mechanical features and genome information, to generate a long-read whole body transcriptome. We deciphered the repeat arrangements of each kind of spidroin, based on which we found that there are substantially transcriptional diversity of each spidroin gene. Some repeat motifs are not documented before. Specifically, we discovered novel full-lengh MaSp transcript as well as a relatively small full-length AcSp isoforms, which are potential promising materials for bioengineering of recombinant spidroin. Our study provided a batch of new spidron resources with detail sequential information. The finding of transcriptional diversity may provide cues in understanding of within-species variation of the mechanical properties of the natural spider silk and further molecular designing of recombinant spidroin.


Subject(s)
Fibroins/chemistry , Fibroins/genetics , Spiders/genetics , Amino Acid Sequence , Animals , China , Evolution, Molecular , Gene Expression Profiling/methods , Genetic Variation/genetics , Phylogeny , Sequence Analysis , Silk/chemistry , Transcriptome/genetics
8.
Am J Gastroenterol ; 115(11): 1891-1901, 2020 11.
Article in English | MEDLINE | ID: mdl-33156108

ABSTRACT

INTRODUCTION: Duodenal epithelial barrier impairment and immune activation may play a role in the pathogenesis of functional dyspepsia (FD). This study was aimed to evaluate the duodenal epithelium of patients with FD and healthy individuals for detectable microscopic structural abnormalities. METHODS: This is a prospective study using esophagogastroduodenoscopy enhanced with duodenal confocal laser endomicroscopy (CLE) and mucosal biopsies in patients with FD (n = 16) and healthy controls (n = 18). Blinded CLE images analysis evaluated the density of epithelial gaps (cell extrusion zones), a validated endoscopic measure of the intestinal barrier status. Analyses of the biopsied duodenal mucosa included standard histology, quantification of mucosal immune cells/cytokines, and immunohistochemistry for inflammatory epithelial cell death called pyroptosis. Transepithelial electrical resistance (TEER) was measured using Ussing chambers. Epithelial cell-to-cell adhesion proteins expression was assessed by real-time polymerase chain reaction. RESULTS: Patients with FD had significantly higher epithelial gap density on CLE in the distal duodenum than that of controls (P = 0.002). These mucosal abnormalities corresponded to significant changes in the duodenal biopsy samples of patients with FD, compared with controls, including impaired mucosal integrity by TEER (P = 0.009) and increased number of epithelial cells undergoing pyroptosis (P = 0.04). Reduced TEER inversely correlated with the severity of certain dyspeptic symptoms. Furthermore, patients with FD demonstrated altered duodenal expression of claudin-1 and interleukin-6. No differences in standard histology were found between the groups. DISCUSSION: This is the first report of duodenal CLE abnormalities in patients with FD, corroborated by biopsy findings of epithelial barrier impairment and increased cell death, implicating that duodenal barrier disruption is a pathogenesis factor in FD and introducing CLE a potential diagnostic biomarker in FD.


Subject(s)
Duodenum/pathology , Dyspepsia/pathology , Endoscopy, Digestive System , Epithelium/pathology , Intestinal Mucosa/pathology , Microscopy, Confocal , Pyroptosis , Adult , Aged , Biopsy , Case-Control Studies , Caspase 1/metabolism , Cell Adhesion/genetics , Claudin-1/genetics , Duodenum/metabolism , Dyspepsia/genetics , Dyspepsia/metabolism , Electric Impedance , Epithelium/metabolism , Female , Humans , Interleukin-6/genetics , Intestinal Mucosa/metabolism , Male , Middle Aged , Young Adult
9.
JCI Insight ; 5(20)2020 10 15.
Article in English | MEDLINE | ID: mdl-33055426

ABSTRACT

High-fat feeding (HFF) leads to gut dysbiosis through unclear mechanisms. We hypothesize that bile acids secreted in response to high-fat diets (HFDs) may act on intestinal Paneth cells, leading to gut dysbiosis. We found that HFF resulted in widespread taxonomic shifts in the bacteria of the ileal mucosa, characterized by depletion of Lactobacillus and enrichment of Akkermansia muciniphila, Clostridium XIVa, Ruminococcaceae, and Lachnospiraceae, which were prevented by the bile acid binder cholestyramine. Immunohistochemistry and in situ hybridization studies showed that G protein-coupled bile acid receptor (TGR5) expressed in Paneth cells was upregulated in the rats fed HFD or normal chow supplemented with cholic acid. This was accompanied by decreased lysozyme+ Paneth cells and α-defensin 5 and 6 and increased expression of XBP-1. Pretreatment with ER stress inhibitor 4PBA or with cholestyramine prevented these changes. Ileal explants incubated with deoxycholic acid or cholic acid caused a decrease in α-defensin 5 and 6 and an increase in XBP-1, which was prevented by TGR5 antibody or 4PBA. In conclusion, this is the first demonstration to our knowledge that TGR5 is expressed in Paneth cells. HFF resulted in increased bile acid secretion and upregulation of TGR5 expression in Paneth cells. Bile acid toxicity in Paneth cells contributes to gut dysbiosis induced by HFF.


Subject(s)
Bile Acids and Salts/metabolism , Dysbiosis/genetics , Gastrointestinal Microbiome/genetics , Receptors, G-Protein-Coupled/genetics , X-Box Binding Protein 1/genetics , Akkermansia/genetics , Akkermansia/pathogenicity , Animals , Bile Acids and Salts/adverse effects , Bile Acids and Salts/biosynthesis , Clostridium/genetics , Clostridium/pathogenicity , Diet, High-Fat/adverse effects , Disease Models, Animal , Dysbiosis/chemically induced , Dysbiosis/metabolism , Dysbiosis/pathology , Gastrointestinal Microbiome/drug effects , Gene Expression Regulation/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Lactobacillus/genetics , Lactobacillus/metabolism , Male , Paneth Cells/metabolism , Paneth Cells/microbiology , Paneth Cells/pathology , Rats , alpha-Defensins/genetics
10.
JCI Insight ; 5(14)2020 07 23.
Article in English | MEDLINE | ID: mdl-32699194

ABSTRACT

The aim of this study was to elucidate the role and the pathways used by bile acid receptor TGR5 in transmitting satiety signals. We showed TGR5 colocalized with cholecystokinin type A (CCK-A) receptors in a subpopulation of rat nodose ganglia (NG) neurons. Intra-arterial injection of deoxycholic acid (DCA) dose-dependently increased firing rate in NG while a subthreshold dose of DCA and CCK-8 increased firing rates synergistically. TGR5-specific agonist oleanolic acid induced NG neuronal firing in a dose-dependent manner. However, the same units did not respond to GW4064, a nuclear receptor-specific agonist. Quantity of DCA-activated neurons in the hypothalamus was determined by c-Fos expression. Combining DCA and CCK-8 caused a 4-fold increase in c-Fos activation. In the arcuate nucleus, c-Fos-positive neurons coexpressed cocaine and amphetamine regulated transcript and proopiomelanocortin. DCA-induced c-Fos expression was eliminated following truncal vagotomy or silencing of TGR5 in the NG. Feeding studies showed intravenous injection of 1 µg/kg of DCA reduced food intake by 12% ± 3%, 24% ± 5%, and 32% ± 6% in the first 3 hours, respectively. Silencing of TGR5 or CCK-A receptor in the NG enhanced spontaneous feeding by 18% ± 2% and 13.5% ± 2.4%, respectively. When both TGR5 and CCK-A receptor were silenced, spontaneous feeding was enhanced by 37% ± 4% in the first 3 hours, suggesting that bile acid may have a physiological role in regulating satiety. Working in concert with CCK, bile acid synergistically enhanced satiety signals to reduce spontaneous feeding.


Subject(s)
Bile Acids and Salts/pharmacology , Deoxycholic Acid/pharmacology , Neurons/drug effects , Receptor, Cholecystokinin A/genetics , Receptors, G-Protein-Coupled/genetics , Afferent Pathways/drug effects , Animals , Bile Acids and Salts/metabolism , Gene Expression Regulation/drug effects , Humans , Isoxazoles/pharmacology , Leptin/genetics , Neurons/pathology , Nodose Ganglion/drug effects , Rats , Receptor, Cholecystokinin A/antagonists & inhibitors , Satiety Response/drug effects , Satiety Response/physiology , Vagus Nerve/drug effects , Vagus Nerve/pathology
11.
J Glob Antimicrob Resist ; 19: 192-193, 2019 12.
Article in English | MEDLINE | ID: mdl-31520808

ABSTRACT

OBJECTIVES: The rapid spread of Klebsiella spp. is recognised as a major threat to public health owing to a rise in the number both of healthcare- and community-acquired infections. Here we report the draft genome sequence of a high carbapenem-resistant Klebsiella quasipneumoniae subsp. quasipneumoniae strain (Cln185) isolated from a human immunodeficiency virus (HIV)-positive patient with pneumonia. METHODS: Classical microbiological methods were applied to isolate and identify the strain. Genomic DNA was sequenced using an Illumina HiSeq platform and the reads were de novo assembled into contigs using CLC Genomics Workbench. The assembled contigs was annotated and whole-genome sequencing (WGS) was performed. RESULTS: WGS analysis revealed that the genome comprised a circular chromosome of 5 406 774bp with a GC content of 57.73%. Three important antimicrobial resistance genes (blaIMP-38, blaOKP-B-6 and blaDHA-1) were detected. In addition, genes conferring resistance to aminoglycosides, ß-lactams, fluoroquinolones and tetracycline were also identified. CONCLUSION: The draft genome sequence reported here will lay the foundation for future research on antimicrobial resistance and pathogenic mechanisms in K. quasipneumoniae subsp. quasipneumoniae and also will promote comparative analysis with genomic features among different sources of clinically important multidrug-resistant strains.


Subject(s)
Carbapenems/pharmacology , Drug Resistance, Multiple, Bacterial , HIV Infections/microbiology , Klebsiella/genetics , Pneumonia/microbiology , Whole Genome Sequencing/methods , Adult , Base Composition , Coinfection , Genome Size , Genome, Bacterial , High-Throughput Nucleotide Sequencing , Humans , Klebsiella/isolation & purification , Male , Microbial Sensitivity Tests
12.
Polymers (Basel) ; 11(5)2019 May 07.
Article in English | MEDLINE | ID: mdl-31067705

ABSTRACT

This work provides a simple method for the preparation of thermoplastic chitosan using the most common dilute inorganic and organic acids in aqueous solutions, namely hydrochloric acid (HCl) and acetic acid (HAc). The melting plasticization behavior of chitosan under different concentrations and types of acid solution was investigated. By means of infrared spectra (IR), scanning electron microscope (SEM), X-ray diffraction (XRD), and other characterization methods, as well as a mechanical property test, it was found that as the acid solution concentration increased, the protonation effect was stronger and the plasticization performance showed a better trend. The structure and performance of the modified chitosan were optimal when the concentration of HCl was around 8 wt %. In addition, it was found that HCl had a better effect on the plasticization of chitosan than HAc, which was because the protonation ability of HCl was stronger than that of HAc. Unlike the casting method, the structure and properties of chitosan sheets prepared by thermoplastic processing were directly affected by protonation, however not by the interaction of anionic-cationic electrostatic attractions between the -NH3+ groups of chitosan chains and the carboxyl groups of acetic acids or the chloridoid groups of hydrochloric acid.

13.
J Neurophysiol ; 121(3): 928-939, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30649980

ABSTRACT

Hyperphagia is common in diabetes and may worsen hyperglycemia and diabetic complications. The responsible mechanisms are not well understood. The hypothalamus is a key center for the control of appetite and energy homeostasis. The ventromedial nucleus (VMH) and arcuate nucleus (ARC) are two critical nuclei involved in these processes. We have reported that R-spondin 1 (Rspo1) and its receptor leucin-rich repeat and G protein-coupled receptor 4 (LGR4) in the VMH and ARC suppressed appetite, but the downstream neuronal pathways are unclear. Here we show that neurons containing cocaine and amphetamine-regulated transcript (CART) in ARC express both LGR4 and insulin receptor; intracerebroventricular injection of Rspo1 induced c-Fos expression in CART neurons of ARC; and silencing CART in ARC attenuated the anorexigenic actions of Rspo1. In diabetic and obese fa/fa rats, Rspo1 mRNA in VMH and CART mRNA in ARC were reduced; this was accompanied by increased food consumption. Insulin treatment restored Rspo1 and CART gene expressions and normalized eating behavior. Chronic intracerebroventricular injection of Rspo1 inhibited food intake and normalized diabetic hyperphagia; intracerebroventricular injection of Rspo1 or insulin increased CART mRNA in ARC. In the CART neuron cell line, Rspo1 and insulin potentiated each other on pERK and ß-catenin, and in rats, they acted synergistically to inhibit food intake. Silencing Rspo1 in VMH reduced CART expression in ARC and attenuated the inhibitory effect of insulin on food intake. In conclusion, our data indicated that CART works downstream of Rspo1 and Rspo1 mediated the action of insulin centrally. The altered Rspo1/CART neurocircuit in the hypothalamus contributes to hyperphagia in diabetes. NEW & NOTEWORTHY This study reports that cocaine and amphetamine-regulated transcript (CART) neurons in the arcuate nucleus (ARC) of hypothalamus acted downstream of R-spondin 1 (Rspo1) to inhibit food intake. The Rspo1 mRNA level in ventromedial nucleus (VMH) and CART mRNA level in ARC were reduced in type 1 diabetic rat and obese fa/fa rat. Rspo1 and insulin acted synergistically on phospho-ERK and ß-catenin signal pathways and in suppressing food intake. The current results proposed that altered Rspo1/CART neurocircuit in the hypothalamus contributes to hyperphagia in diabetes.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Hyperphagia/metabolism , Hypothalamus/metabolism , Nerve Tissue Proteins/metabolism , Thrombospondins/metabolism , Animals , Cell Line , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/physiopathology , Eating/drug effects , Hyperphagia/drug therapy , Hyperphagia/etiology , Hyperphagia/physiopathology , Hypothalamus/physiopathology , Insulin/pharmacology , Insulin/therapeutic use , Male , Mice , Nerve Tissue Proteins/genetics , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Thrombospondins/genetics
14.
J Clin Invest ; 128(1): 267-280, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29202473

ABSTRACT

Foods high in fermentable oligosaccharides, disaccharides, monosaccharides, and polyols (FODMAPs) exacerbate symptoms of irritable bowel syndrome (IBS); however, their mechanism of action is unknown. We hypothesized that a high-FODMAP (HFM) diet increases visceral nociception by inducing dysbiosis and that the FODMAP-altered gut microbial community leads to intestinal pathology. We fed rats an HFM and showed that HFM increases rat fecal Gram-negative bacteria, elevates lipopolysaccharides (LPS), and induces intestinal pathology, as indicated by inflammation, barrier dysfunction, and visceral hypersensitivity (VH). These manifestations were prevented by antibiotics and reversed by low-FODMAP (LFM) diet. Additionally, intracolonic administration of LPS or fecal supernatant (FS) from HFM-fed rats caused intestinal barrier dysfunction and VH, which were blocked by the LPS antagonist LPS-RS or by TLR4 knockdown. Fecal LPS was higher in IBS patients than in healthy subjects (HS), and IBS patients on a 4-week LFM diet had improved IBS symptoms and reduced fecal LPS levels. Intracolonic administration of FS from IBS patients, but not FS from HS or LFM-treated IBS patients, induced VH in rats, which was ameliorated by LPS-RS. Our findings indicate that HFM-associated gut dysbiosis and elevated fecal LPS levels induce intestinal pathology, thereby modulating visceral nociception and IBS symptomatology, and might provide an explanation for the success of LFM diet in IBS patients.


Subject(s)
Dietary Carbohydrates/adverse effects , Dysbiosis , Gastrointestinal Microbiome , Intestines/microbiology , Irritable Bowel Syndrome , Lipopolysaccharides/toxicity , Nociception , Animals , Dietary Carbohydrates/pharmacology , Dysbiosis/chemically induced , Dysbiosis/genetics , Dysbiosis/metabolism , Dysbiosis/microbiology , Gene Knockdown Techniques , Irritable Bowel Syndrome/chemically induced , Irritable Bowel Syndrome/genetics , Irritable Bowel Syndrome/metabolism , Irritable Bowel Syndrome/microbiology , Male , Rats , Rats, Wistar , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
15.
Gastroenterology ; 151(5): 910-922.e7, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27475306

ABSTRACT

BACKGROUND & AIMS: Patients with diabetes have defects in the vagal afferent pathway that result in abnormal gastrointestinal function. We investigated whether selective increased activation of the 2-pore domain potassium channel TRESK (2-pore-domain weak inward-rectifying potassium channel-related spinal cord potassium channel) contributes to nodose ganglia (NG) malfunction, disrupting gastrointestinal function in diabetic rats. METHODS: We conducted whole-cell current-clamp and single-unit recordings in NG neurons from diabetes-prone BioBreeding/Worcester rats and streptozotocin-induced diabetic (STZ-D) rats and compared them with control rats. NG neurons in rats or cultured NG neurons were exposed to pharmacologic antagonists and/or transfected with short hairpin or small interfering RNAs that reduced expression of TRESK. We then made electrophysiologic recordings and studied gastrointestinal functions. RESULTS: We observed reduced input resistance, hyperpolarized membrane potential, and increased current threshold to elicit action potentiation in NG neurons of STZ-D rats compared with controls. NG neuron excitability was similarly altered in diabetes-prone rats. In vivo single-unit NG neuronal discharges in response to 30 and 60 pmol cholecystokinin octapeptide were significantly lower in STZ-D rats compared with controls. Reducing expression of the TRESK K+ channel restored NG excitability in vitro and in vivo, as well as cholecystokinin 8-stimulated secretion of pancreatic enzymes and secretin-induced gastrointestinal motility, which are mediated by vago-vagal reflexes. These abnormalities resulted from increased intracellular Ca2+ in the NG, activating calcineurin, which, in turn, bound to an nuclear factor of activated T cell-like docking site on the TRESK protein, resulting in neuronal membrane hyperpolarization. CONCLUSIONS: In 2 rate models of diabetes, we found that activation of the TRESK K+ channel reduced NG excitability and disrupted gastrointestinal functions.


Subject(s)
Diabetes Mellitus, Experimental/physiopathology , Gastrointestinal Motility/physiology , Nodose Ganglion/physiopathology , Potassium Channels/metabolism , Animals , Biomarkers/metabolism , Diabetes Mellitus, Experimental/metabolism , Male , Membrane Potentials , Patch-Clamp Techniques , Rats , Rats, Inbred BB , Reflex
16.
J Physiol ; 593(17): 3973-89, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26174421

ABSTRACT

Ghrelin, a hunger signalling peptide derived from the peripheral tissues, overcomes the satiety signals evoked by anorexigenic molecules, such as cholecystokinin (CCK) and leptin, to stimulate feeding. Using in vivo and in vitro electrophysiological techniques, we show that ghrelin hyperpolarizes neurons and inhibits currents evoked by leptin and CCK-8. Administering a KATP channel antagonist or silencing Kir6.2, a major subunit of the KATP channel, abolished ghrelin inhibition. The inhibitory actions of ghrelin were also abolished by treating the vagal ganglia neurons with pertussis toxin, as well as phosphatidylinositol 3-kinase (PI3K) or extracellular signal-regulated kinase 1 and 2 (Erk1/2) small interfering RNA. Feeding experiments showed that silencing Kir6.2 in the vagal ganglia abolished the orexigenic actions of ghrelin. These data indicate that ghrelin modulates vagal ganglia neuron excitability by activating KATP conductance via the growth hormone secretagogue receptor subtype 1a-Gαi -PI3K-Erk1/2-KATP pathway. This provides a mechanism to explain the actions of ghrelin with respect to overcoming anorexigenic signals that act via the vagal afferent pathways. Ghrelin is the only known hunger signal derived from the peripheral tissues. Ghrelin overcomes the satiety signals evoked by anorexigenic molecules, such as cholecystokinin (CCK) and leptin, to stimulate feeding. The mechanisms by which ghrelin reduces the sensory signals evoked by anorexigenic hormones, which act via the vagus nerve to stimulate feeding, are unknown. Patch clamp recordings of isolated rat vagal neurons show that ghrelin hyperpolarizes neurons by activating K(+) conductance. Administering a KATP channel antagonist or silencing Kir6.2, a major subunit of the KATP channel, abolished ghrelin inhibition in vitro and in vivo. Patch clamp studies show that ghrelin inhibits currents evoked by leptin and CCK-8, which operate through independent ionic channels. The inhibitory actions of ghrelin were abolished by treating the vagal ganglia neurons with pertussis toxin, as well as phosphatidylinositol 3-kinase (PI3K) or extracellular signal-regulated kinase 1 and 2 (Erk1/2) small interfering RNA. In vivo gene silencing of PI3K and Erk1/2 in the nodose ganglia prevented ghrelin inhibition of leptin- or CCK-8-evoked vagal firing. Feeding experiments showed that silencing Kir6.2 in the vagal ganglia abolished the orexigenic actions of ghrelin. These data indicate that ghrelin modulates vagal ganglia neuron excitability by activating KATP conductance via the growth hormone secretagogue receptor subtype 1a-Gαi -PI3K-Erk1/2-KATP pathway. The resulting hyperpolarization renders the neurons less responsive to signals evoked by anorexigenic hormones. This provides a mechanism to explain the actions of ghrelin with respect to overcoming anorexigenic signals that act via the vagal afferent pathways.


Subject(s)
Ghrelin/pharmacology , KATP Channels/physiology , Nodose Ganglion/physiology , Sensory Receptor Cells/physiology , Animals , Cholecystokinin/pharmacology , Eating , KATP Channels/antagonists & inhibitors , KATP Channels/genetics , Leptin/pharmacology , Male , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Mitogen-Activated Protein Kinase 3/physiology , Nodose Ganglion/drug effects , Peptide Fragments/pharmacology , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinases/physiology , Potassium Channel Blockers/pharmacology , RNA, Small Interfering/genetics , Rats, Sprague-Dawley , Sensory Receptor Cells/drug effects
17.
Endocrinology ; 154(1): 296-307, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23211706

ABSTRACT

Glucosensing nodose ganglia neurons mediate the effects of hyperglycemia on gastrointestinal motility. We hypothesized that the glucose-sensing mechanisms in the nodose ganglia are similar to those of hypothalamic glucose excited neurons, which sense glucose through glycolysis. Glucose metabolism leads to ATP-sensitive potassium channel (K(ATP)) channel closure and membrane depolarization. We identified glucosensing elements in the form of glucose transporters (GLUTs), glucokinase (GK), and K(ATP) channels in rat nodose ganglia and evaluated their physiological significance. In vitro stomach-vagus nerve preparations demonstrated the gastric vagal afferent response to elevated glucose. Western blots and RT-PCR revealed the presence of GLUT1, GLUT3, GLUT4, GK, and Kir6.2 in nodose ganglia neurons and gastric branches of the vagus nerve. Immunocytochemistry confirmed the expression of GLUT3, GK, and Kir6.2 in nodose ganglia neurons (46.3 ± 3%). Patch-clamp studies detected glucose excitation in 30% (25 of 83) of gastric-projecting nodose ganglia neurons, which was abolished by GLUT3 or GK short hairpin RNA transfections. Silencing GLUT1 or GLUT4 in nodose ganglia neurons did not prevent the excitatory response to glucose. Elevated glucose elicited a response from 43% of in vitro nerve preparations. A dose-dependent response was observed, reaching maximum at a glucose level of 250 mg/dl. The gastric vagal afferent responses to glucose were inhibited by diazoxide, a K(ATP) channel opener. In conclusion, a subset of neurons in the nodose ganglia and gastric vagal afferents are glucoresponsive. Glucosensing requires a GLUT, GK, and K(ATP) channels. These elements are transported axonally to the gastric vagal afferents, which can be activated by elevated glucose through modulation of K(ATP) channels.


Subject(s)
Electrophysiology/methods , Gastric Mucosa/metabolism , Immunohistochemistry/methods , Nodose Ganglion/metabolism , Stomach/innervation , Vagus Nerve/metabolism , Animals , Blotting, Western , Diazoxide/pharmacology , Glucokinase/metabolism , Glucose/pharmacology , Glucose Transport Proteins, Facilitative/metabolism , KATP Channels/metabolism , Male , Nodose Ganglion/drug effects , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Stomach/drug effects , Vagus Nerve/drug effects
18.
Am J Physiol Gastrointest Liver Physiol ; 303(9): G1042-51, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22936273

ABSTRACT

Vagal CCK-A receptors (CCKARs) and leptin receptors (LRbs) interact synergistically to mediate short-term satiety. Cocaine- and amphetamine-regulated transcript (CART) peptide is expressed by vagal afferent neurons. We sought to demonstrate that this neurotransmitter regulates CCK and leptin actions on short-term satiety. We also examined the signal transduction pathways responsible for mediating the CART release from the nodose ganglia (NG). ELISA studies coupled with gene silencing of NG neurons by RNA interference elucidated intracellular signaling pathways responsible for CCK/leptin-stimulated CART release. Feeding studies followed by gene silencing of CART in NG established the role of CART in mediating short-term satiety. Immunohistochemistry was performed on rat NG neurons to confirm colocalization of CCKARs and LRbs; 63% of these neurons contained CART. Coadministration of CCK-8 and leptin caused a 2.2-fold increase in CART release that was inhibited by CCK-OPE, a low-affinity CCKAR antagonist. Transfection of cultured NG neurons with steroid receptor coactivator (SRC) or phosphatidylinositol 3-kinase (PI3K) small-interfering RNA (siRNA) or STAT3 lentiviral short hairpin RNA inhibited CCK/leptin-stimulated CART release. Silencing the expression of the EGR-1 gene inhibited the CCK/leptin-stimulated CART release but had no effect on CCK/leptin-stimulated neuronal firing. Electroporation of NG with CART siRNA inhibited CCK/leptin stimulated c-Fos expression in rat hypothalamus. Feeding studies following electroporation of the NG with CART or STAT3 siRNA abolished the effects of CCK/leptin on short-term satiety. We conclude that the synergistic interaction of low-affinity vagal CCKARs and LRbs mediates CART release from the NG, and CART is the principal neurotransmitter mediating short-term satiety. CART release from the NG involves interaction between CCK/SRC/PI3K cascades and leptin/JAK2/PI3K/STAT3 signaling pathways.


Subject(s)
Leptin , Nerve Tissue Proteins , Nodose Ganglion , Satiation/drug effects , Sincalide , Animals , Down-Regulation , Electroporation , Gene Silencing , Immunohistochemistry , Leptin/metabolism , Leptin/pharmacology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nodose Ganglion/drug effects , Nodose Ganglion/metabolism , Nuclear Receptor Coactivators/metabolism , RNA, Small Interfering/metabolism , Rats , Receptor, Cholecystokinin A/metabolism , Receptors, Leptin/metabolism , Sincalide/metabolism , Sincalide/pharmacology , Synaptic Transmission/genetics
20.
Am J Physiol Gastrointest Liver Physiol ; 300(3): G394-400, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21193530

ABSTRACT

The inhibitory action of hyperglycemia is mediated by vagal afferent fibers innervating the stomach and duodenum. Our in vitro studies showed that a subset of nodose ganglia neurons is excited by rising ambient glucose, involving inactivation of ATP-sensitive K(+) (K(ATP)) channels and leading to membrane depolarization and neuronal firing. To investigate whether nodose ganglia K(ATP) channels mediate gastric relaxation induced by hyperglycemia, we performed in vivo gastric motility studies to examine the effects of K(ATP) channel activators and inactivators. Intravenous infusion of 20% dextrose induced gastric relaxation in a dose-dependent manner. This inhibitory effect of hyperglycemia was blocked by diazoxide, a K(ATP) channel activator. Conversely, tolbutamide, a K(ATP) channel inactivator, induced dose-dependent gastric relaxation, an effect similar to hyperglycemia. Vagotomy, perivagal capsaicin treatment, and hexamethonium each prevented the inhibitory action of tolbutamide. Similarly, N(G)-nitro-l-arginine methyl ester, an inhibitor of nitric oxide synthase, also blocked tolbutamide's inhibitory effect. To show that K(ATP) channel inactivation at the level of the nodose ganglia induces gastric relaxation, we performed electroporation of the nodose ganglia with small interfering RNA of Kir6.2 (a subunit of K(ATP)) and plasmid pEGFP-N1 carrying the green fluorescent protein gene. The gastric responses to hyperglycemia and tolbutamide were not observed in rats with Kir6.2 small interfering RNA-treated nodose ganglia. However, these rats responded to secretin, which acts via the vagal afferent pathway, independently of K(ATP) channels. These studies provide in vivo evidence that hyperglycemia induces gastric relaxation via the vagal afferent pathway. This action is mediated through inactivation of nodose ganglia K(ATP) channels.


Subject(s)
Gastrointestinal Motility , Gastroparesis/etiology , Hyperglycemia/complications , KATP Channels/metabolism , Muscle Relaxation , Nodose Ganglion/metabolism , Stomach/innervation , Animals , Blotting, Western , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Ganglionic Blockers/pharmacology , Gastrointestinal Motility/drug effects , Gastroparesis/metabolism , Gastroparesis/physiopathology , Gastroparesis/prevention & control , Glucose/administration & dosage , Hyperglycemia/metabolism , Hyperglycemia/physiopathology , Immunohistochemistry , Infusions, Intravenous , KATP Channels/drug effects , KATP Channels/genetics , Male , Muscle Relaxation/drug effects , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Nodose Ganglion/drug effects , Nodose Ganglion/physiopathology , Potassium Channel Blockers/pharmacology , Potassium Channels, Inwardly Rectifying/genetics , Potassium Channels, Inwardly Rectifying/metabolism , RNA Interference , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Sensory System Agents/pharmacology , Stomach/drug effects , Vagotomy
SELECTION OF CITATIONS
SEARCH DETAIL
...