Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Mol Cell Neurosci ; 126: 103873, 2023 09.
Article in English | MEDLINE | ID: mdl-37295578

ABSTRACT

A relatively new pharmacological target in obesity treatment has been the preproglucagon (PPG) signalling, predominantly with glucagon-like peptide (GLP) 1 receptor agonists. As far as the PPG role within the digestive system is well recognised, its actions in the brain remain understudied. Here, we investigated PPG signalling in the Dorsomedial Hypothalamus (DMH), a structure involved in feeding regulation and metabolism, using in situ hybridisation, electrophysiology, and immunohistochemistry. Our experiments were performed on animals fed both control, and high-fat diet (HFD), uncovering HFD-mediated alterations. First, sensitivity to exendin-4 (Exn4, a GLP1R agonist) was shown to increase under HFD, with a higher number of responsive neurons. The amplitude of the response to both Exn4 and oxyntomodulin (Oxm) was also altered, diminishing its relationship with the cells' spontaneous firing rate. Not only neuronal sensitivity, but also GLP1 presence, and therefore possibly release, was influenced by HFD. Immunofluorescent labelling of the GLP1 showed changes in its density depending on the metabolic state (fasted/fed), but this effect was eliminated by HFD feeding. Interestingly, these dietary differences were absent after a period of restricted feeding, allowing for an anticipation of the alternating metabolic states, which suggests possible prevention of such outcome.


Subject(s)
Diet, High-Fat , Hypothalamus , Proglucagon , Signal Transduction , Animals , Rats , Hypothalamus/physiology , Proglucagon/metabolism , Rats, Sprague-Dawley , Male , Glucagon-Like Peptide-1 Receptor/genetics , Glucagon-Like Peptide-1 Receptor/metabolism , Glucagon-Like Peptide-2 Receptor/genetics , Glucagon-Like Peptide-2 Receptor/metabolism , RNA, Messenger/metabolism , Neurons/metabolism , Synapses , Nerve Fibers/metabolism , Electrophysiology , Proto-Oncogene Proteins c-fos/metabolism , Satiety Response , Feeding Behavior
2.
Nat Metab ; 4(10): 1402-1419, 2022 10.
Article in English | MEDLINE | ID: mdl-36266547

ABSTRACT

The hypothalamus plays a key role in coordinating fundamental body functions. Despite recent progress in single-cell technologies, a unified catalog and molecular characterization of the heterogeneous cell types and, specifically, neuronal subtypes in this brain region are still lacking. Here, we present an integrated reference atlas, 'HypoMap,' of the murine hypothalamus, consisting of 384,925 cells, with the ability to incorporate new additional experiments. We validate HypoMap by comparing data collected from Smart-Seq+Fluidigm C1 and bulk RNA sequencing of selected neuronal cell types with different degrees of cellular heterogeneity. Finally, via HypoMap, we identify classes of neurons expressing glucagon-like peptide-1 receptor (Glp1r) and prepronociceptin (Pnoc), and validate them using single-molecule in situ hybridization. Collectively, HypoMap provides a unified framework for the systematic functional annotation of murine hypothalamic cell types, and it can serve as an important platform to unravel the functional organization of hypothalamic neurocircuits and to identify druggable targets for treating metabolic disorders.


Subject(s)
Glucagon-Like Peptide-1 Receptor , Hypothalamus , Mice , Animals , Glucagon-Like Peptide-1 Receptor/genetics , Hypothalamus/metabolism , Neurons/metabolism , Sequence Analysis, RNA , Gene Expression
3.
Appetite ; 174: 106022, 2022 07 01.
Article in English | MEDLINE | ID: mdl-35430298

ABSTRACT

OBJECTIVE: The hypothalamus is a key region of the brain implicated in homeostatic regulation, and is an integral centre for the control of feeding behaviour. Glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) are incretin hormones with potent glucoregulatory function through engagement of their respective cognate receptors, GLP-1R and GIPR. Recent evidence indicates that there is a synergistic effect of combining GIP- and GLP-1-based pharmacology on appetite and body weight. The mechanisms underlying the enhanced weight loss exhibited by GIPR/GLP-1R co-agonism are unknown. Gipr and Glp1r are expressed in the hypothalamus in both rodents and humans. To better understand incretin receptor-expressing cell populations, we compared the cell types and expression profiles of Gipr- and Glp1r-expressing hypothalamic cells using single-cell RNA sequencing. METHODS: Using Glp1r-Cre or Gipr-Cre transgenic mouse lines, fluorescent reporters were introduced into either Glp1r- or Gipr-expressing cells, respectively, upon crossing with a ROSA26-EYFP reporter strain. From the hypothalami of these mice, fluorescent Glp1rEYFP+ or GiprEYFP+ cells were FACS-purified and sequenced using single-cell RNA sequencing. Transcriptomic analysis provided a survey of both non-neuronal and neuronal cells, and comparisons between Glp1rEYFP+ and GiprEYFP + populations were made. RESULTS: A total of 14,091 Glp1rEYFP+ and GiprEYFP+ cells were isolated, sequenced and taken forward for bioinformatic analysis. Both Glp1rEYFP+ and GiprEYFP+ hypothalamic populations were transcriptomically highly heterogeneous, representing vascular cell types, oligodendrocytes, astrocytes, microglia, and neurons. The majority of GiprEYFP+ cells were non-neuronal, whereas the Glp1rEYFP+ population was evenly split between neuronal and non-neuronal cell types. Both Glp1rEYFP+ and GiprEYFP+ oligodendrocytes express markers for mature, myelin-forming oligodendrocytes. While mural cells are represented in both Glp1rEYFP+ and GiprEYFP+ populations, Glp1rEYFP+ mural cells are largely smooth muscle cells, while the majority of GiprEYFP+ mural cells are pericytes. The co-expression of regional markers indicate that clusters of Glp1rEYFP+ and GiprEYFP+ neurons have been isolated from the arcuate, ventromedial, lateral, tuberal, suprachiasmatic, and premammillary nuclei of the hypothalamus. CONCLUSIONS: We have provided a detailed comparison of Glp1r and Gipr cells of the hypothalamus with single-cell resolution. This resource will provide mechanistic insight into how engaging Gipr- and Glp1r-expressing cells of the hypothalamus may result in changes in feeding behaviour and energy balance.


Subject(s)
Glucagon-Like Peptide-1 Receptor , Incretins , Animals , Gastric Inhibitory Polypeptide/genetics , Gastric Inhibitory Polypeptide/metabolism , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor/genetics , Glucagon-Like Peptide-1 Receptor/metabolism , Glucose , Humans , Hypothalamus/metabolism , Mice , Transcriptome
4.
J Agric Food Chem ; 69(1): 212-222, 2021 Jan 13.
Article in English | MEDLINE | ID: mdl-33353303

ABSTRACT

ι-Carrageenan performs diversified biological activities but has low bioavailability. ι-Carrageenan tetrasaccharide (ιCTs), a novel marine oligosaccharide prepared by the marine enzyme Cgi82A, was investigated for its effects on insulin resistance in high-fat and high-sucrose diet mice. Oral administration of ιCTs (ιCTs-L 30.0 mg/kg·bw, ιCTs-H 90.0 mg/kg·bw) decreased fasting blood glucose by 35.1% ± 1.41 (P < 0.01) and 27.4% ± 0.420 (P < 0.05), and enhanced glucose tolerance. Besides, ιCTs-L ameliorated islet vacuolization, decreased the ß cell apoptosis by 21.8% ± 0.200 (P < 0.05), and promoted insulin secretion by 5.41% ± 0.0173 (P < 0.01) through pancreatic hematoxylin and eosin (H&E) staining, TUNEL staining, and insulin-glucagon immunostaining analysis. Interestingly, ιCTs-L and ιCTs-H treatment increased the incretin GLP-1 content in serum by 22.1% ± 0.402 (P < 0.01) and 10.7% ± 0.0935 (P < 0.05) respectively, through regulating the bile acid levels, which contributed to the inhibition of ß cell apoptosis. Mechanically, ιCTs upregulated the expression of the GLP-1 receptor (GLP-1R) and protein kinase A (PKA) in the GLP-1/cAMP/PKA signaling pathway, and further inhibited the expression of cytochrome C and caspase 3 in the mitochondrial apoptotic pathway. In conclusion, this study suggested that ιCTs alleviated insulin resistance by GLP-1-mediated inhibition of ß cell apoptosis and proposed a new strategy for developing potential functional foods that prevent insulin resistance.


Subject(s)
Apoptosis/drug effects , Carrageenan/administration & dosage , Diabetes Mellitus, Type 2/drug therapy , Glucagon-Like Peptide 1/metabolism , Insulin-Secreting Cells/cytology , Mitochondria/drug effects , Oligosaccharides/administration & dosage , Plant Extracts/administration & dosage , Animals , Blood Glucose/metabolism , Carrageenan/chemistry , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/physiopathology , Glucagon-Like Peptide 1/genetics , Glucagon-Like Peptide-1 Receptor/genetics , Glucagon-Like Peptide-1 Receptor/metabolism , Humans , Insulin Resistance , Insulin Secretion/drug effects , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Oligosaccharides/analysis , Plant Extracts/analysis , Signal Transduction/drug effects
5.
Nutrients ; 12(10)2020 Oct 08.
Article in English | MEDLINE | ID: mdl-33049944

ABSTRACT

Fucoidan extracted from brown algae has multiple beneficial functions. In this study, we investigated the effects of low-molecular-weight fucoidan (oligo-FO) on renal fibrosis under in vitro and in vivo diabetic conditions, and its molecular mechanisms. Advanced glycation product (AGE)-stimulated rat renal proximal tubular epithelial cells (NRK-52E) and diabetic mice induced by high-fat diet and intraperitoneal injection of streptozotocin and nicotinamide were used. Oligo-FO treatment significantly inhibited anti-high mobility group box 1 (HMGB1)/RAGE/ anti-nuclear factor-kappa B (NF-κB)/transforming growth factor-ß1 (TGF-ß1)/TGF-ß1R/Smad 2/3/fibronectin signaling pathway and HIF-1α activation in AGE-stimulated NRK-52E cells. Conversely, the expression and activity of Sirt-1; the levels of ubiquitin-specific peptidase 22 (USP22), p-AMPK, glucagon-like peptide-1 receptor (GLP-1R), and heme oxygenase-1 (HO-1); and Nrf2 activation were remarkably increased by oligo-FO in AGE-stimulated cells. However, the above effects of oligo-FO were greatly diminished by inhibiting Sirt-1, HO-1, or GLP-1R activity. Similar changes of these pro-fibrotic genes in the kidney and a marked attenuation of renal injury and dysfunction were observed in oligo-FO-treated diabetic mice. These findings indicated that the inhibitory effects of the oligo-FO on diabetes-evoked renal fibrosis are mediated by suppressing TGF-ß1-activated pro-fibrogenic processes via Sirt-1, HO-1, and GLP-1R dependence. Collectively, fucoidan-containing foods or supplements may be potential agents for ameliorating renal diseases due to excessive fibrosis.


Subject(s)
Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/genetics , Dietary Supplements , Gene Expression/drug effects , Glucagon-Like Peptide-1 Receptor/genetics , Glucagon-Like Peptide-1 Receptor/metabolism , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Kidney/pathology , Membrane Proteins/genetics , Membrane Proteins/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Nutritional Physiological Phenomena/physiology , Phaeophyceae/chemistry , Phytotherapy , Polysaccharides/pharmacology , Sirtuin 1/genetics , Sirtuin 1/metabolism , Animals , Cells, Cultured , Fibrosis/drug therapy , Fibrosis/genetics , Male , Mice, Inbred C57BL , Molecular Weight , Polysaccharides/administration & dosage , Polysaccharides/chemistry , Polysaccharides/isolation & purification , Rats
6.
Biomed Res Int ; 2019: 8724824, 2019.
Article in English | MEDLINE | ID: mdl-31828140

ABSTRACT

BACKGROUND: Glucagon-like peptide 1 (GLP-1) hormone is an incretin hormone that is secreted in the ileum and plays a role in the pancreas to increase insulin secretion, stimulate proliferation, and prevent pancreatic ß-cell apoptosis. Currently, diabetes mellitus (DM) treatment based on GLP-1 work is being developed, for instance, from herbal plants such as Hibiscus sabdariffa Linn (H. sabdariffa). Therefore, this study aims to determine the potential of H. sabdariffa in GLP-1 secretion in the ileum and its action in pancreatic ß-cells. In addition, this study also aims to determine the active ingredients of H. sabdariffa (Hib) that interact with sodium-glucose cotransporter-1 (SGLT-1) so that it can increase GLP-1 secretion in the ileum and interact with GLP-1 receptors (GLP-1R) in the pancreas. METHOD: This experimental study used 24 experimental animals of Sprague-Dawley type (aged 8-10 weeks, weight 200-250 g) that were divided into 6 groups, namely, (i) normal (C), (ii) normal-Hib 200 (C-Hib200), (iii) normal-Hib 500 (C-Hib500), (iv) DM (C-DM), (v) DM-Hib200, and (vi) DM-Hib500. H. sabdariffa extract was given orally once a day for 5 weeks. Testing of GLP-1 levels in the ileum and pancreatic tissue was performed by enzyme-linked immunosorbent assay. The prediction of the interaction mechanism of the active substance H. sabdariffa against GLP-1 was done using molecular docking. RESULTS: There was a decrease in GLP-1 levels in the ileum of DM rats (p < 0.05). However, DM rats administered H. sabdariffa 500 mg/kg BW had GLP-1 levels that were the same as in normal rats (p > 0.05). This is due to active ingredients such as leucosin, which binds to SGLT-1. Administration of 500 mg/kg BW H. sabdariffa in DM rats resulted in GLP-1 levels in the pancreas that were the same as in normal rats (p > 0.05). In addition, the active ingredient of H. sabdariffa, delphinidin, binds to GLPR in the pancreas. CONCLUSION: The active ingredient of H. sabdariffa can increase GLP-1 secretion in the ileum and can interact with G protein-linked receptors in the pancreas.


Subject(s)
Diabetes Mellitus/drug therapy , Glucagon-Like Peptide 1/genetics , Glucagon-Like Peptide-1 Receptor/genetics , Hibiscus/chemistry , Sodium-Glucose Transporter 1/genetics , Animals , Apoptosis/drug effects , Blood Glucose/metabolism , Diabetes Mellitus/genetics , Diabetes Mellitus/pathology , Disease Models, Animal , Glucagon-Like Peptide 1/chemistry , Humans , Ileum/metabolism , Ileum/pathology , Incretins/genetics , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Molecular Docking Simulation , Pancreas/drug effects , Pancreas/metabolism , Plant Extracts/pharmacology , Rats , Rats, Sprague-Dawley
7.
Biomolecules ; 9(12)2019 12 12.
Article in English | MEDLINE | ID: mdl-31842341

ABSTRACT

Flavonoids have been shown to modulate GLP-1 in obesity. GLP-1 induces some of its effects through the intestinal GLP-1 receptor (GLP-1R), though no data exist on how flavonoids affect this receptor. Here, we examine how a dose of grape seed proanthocyanidin extract (GSPE) with anti-obesity activity affects intestinal GLP-1R and analyze whether epigenetics play a role in the long-lasting effects of GSPE. We found that 10-day GSPE administration prior to the cafeteria diet upregulated GLP-1R mRNA in the ileum 17 weeks after the GSPE treatment. This was associated with a hypomethylation of the GLP-1R promoter near the region where the SP1 transcription factor binds. In the colon, the cafeteria diet upregulated GLP-1R without showing any GSPE effect. In conclusion, we have identified long-lasting GSPE effects on GLP-1R gene expression in the ileum that are partly mediated by hypomethylation at the gene promoter and may affect the SP1 binding factor.


Subject(s)
DNA Methylation/drug effects , Glucagon-Like Peptide-1 Receptor/genetics , Grape Seed Extract/pharmacology , Ileum/drug effects , Plant Extracts/pharmacology , Proanthocyanidins/pharmacology , Promoter Regions, Genetic/genetics , Up-Regulation/drug effects , Animals , Female , Glucagon-Like Peptide-1 Receptor/metabolism , Grape Seed Extract/administration & dosage , Grape Seed Extract/chemistry , Ileum/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Plant Extracts/administration & dosage , Plant Extracts/chemistry , Proanthocyanidins/administration & dosage , Proanthocyanidins/chemistry , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar
8.
J Food Drug Anal ; 27(1): 135-144, 2019 01.
Article in English | MEDLINE | ID: mdl-30648566

ABSTRACT

Abelmoschus esculentus (AE) has been used in traditional medicine to ameliorate hyperglycemia, but its mucilage increased bioassay difficulties. We have obtained a series of AE subfractions. Among them F1 and F2 regulated dipeptidyl peptidase-4 (DPP-4) and type 1 glucagon-like peptide receptor (GLP-1R), the treatment targets for type 2 diabetes. F1, F2 and fraction residues (FR) showed advantage on different aspects, which attenuates insulin resistance and metabolic disorder in vivo, and prevents renal-tubular change in vitro. In the present study, using type 2 diabetes model induced by high fat diet (HFD) and streptozotocin (STZ), we aim to investigate whether AE prevent diabetic nephropathy by regulating the putative markers. The results showed that all the subfractions ameliorated albuminuria and renal hyperfiltration (measured by creatinine clearance rate; CCr) accompanied with diabetes, while F2 acted most promptly and consistently. Histologically AE reduced renal tubular change, fibrosis and fat deposition. F2 and FR exerted significant effects to decrease DPP-4 while increase GLP-1R. Although all the subfractions were effective to reduce oxidative stress, only F2 acted on kidneys specifically. In conclusion, we have demonstrated AE has benefits to regulate DPP-4 and GLP-1R, to reduce oxidative stress and renal fibrosis, with resultant to improve renal function and prevent diabetic renal damage. Taken together, F2 could be more promising to be developed as adjuvant for diabetic nephropathy.


Subject(s)
Abelmoschus/chemistry , Diabetes Mellitus, Type 2/complications , Diabetic Nephropathies/drug therapy , Dipeptidyl Peptidase 4/metabolism , Dipeptidyl-Peptidase IV Inhibitors/administration & dosage , Drugs, Chinese Herbal/administration & dosage , Glucagon-Like Peptide-1 Receptor/metabolism , Animals , Diabetic Nephropathies/metabolism , Dipeptidyl Peptidase 4/genetics , Glucagon-Like Peptide-1 Receptor/genetics , Humans , Kidney/drug effects , Kidney/metabolism , Male , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley
9.
J Periodontal Res ; 54(2): 106-114, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30207387

ABSTRACT

OBJECTIVE: Liraglutide (LIRA) is a novel antidiabetic therapy that may have anti-inflammatory and bone protective effects. Thus, we studied the potential therapeutic effect of LIRA on periodontitis by assessing the effects of LIRA on the proliferation, migration, inflammation, and osteogenic differentiation of human periodontal ligament cells (hPDLCs) after LPS stimulation. MATERIAL AND METHODS: The expression of glucagon like-peptide 1 receptor (GLP-1R) was measured using qRT-PCR. HPDLCs proliferation after LIRA were analyzed using MTT assays. Cell migration was quantified using a wound-healing assay. The expression of inflammatory (IL-6 and TNF-α) was measured by qRT-PCR and ELISA in hPDLCs. The effect of LIRA on the mineralization potential of hPDLCs was assessed by alizarin red S staining. Furthermore, the expression of Runx2 and ALP was measured by qRT-PCR and Western blot in hPDLCs. RESULTS: GLP-1R mRNA was present on hPDLCs, and LIRA increased the expression of GLP-1R mRNA. When cultured with 25, 50, 75, 100 and 125 nM LIRA for 24 h, hPDLCs proliferation was enhanced in a dose-dependent manner (P < 0.05), and 100 nM was optimal. LIRA promoted hPDLCs migration in a time-dependent manner. LPS significantly increased the expression of IL-6 and TNF-α (P < 0.01), decreased the formation of mineralization nodes (P < 0.01), and inhibited the expression of ALP and Runx2 (P < 0.05). LIRA treatment blocked the expression of IL-6 and TNF-α (P < 0.01), increased the formation of mineralization nodes (P < 0.01), and enhanced the expression of ALP and Runx2 (P < 0.05). CONCLUSION: LIRA can enhance the proliferation, migration, and osteogenic differentiation of hPDLCs and inhibit the inflammatory response. Thus, LIRA may have potential therapeutic use as an adjuvant treatment for human periodontitis, and this effect is independent of hypoglycemic activity.


Subject(s)
Cell Differentiation/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Hypoglycemic Agents/pharmacology , Liraglutide/pharmacology , Osteogenesis/drug effects , Periodontal Ligament/cytology , Periodontitis/pathology , Biomarkers/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Gene Expression , Glucagon-Like Peptide-1 Receptor/genetics , Glucagon-Like Peptide-1 Receptor/metabolism , Humans , Hypoglycemic Agents/therapeutic use , Inflammation , Inflammation Mediators/metabolism , Interleukin-6/metabolism , Liraglutide/therapeutic use , Periodontitis/diagnosis , Periodontitis/drug therapy , Periodontitis/genetics , RNA, Messenger/metabolism , Tumor Necrosis Factor-alpha/metabolism
10.
Mol Metab ; 20: 178-193, 2019 02.
Article in English | MEDLINE | ID: mdl-30528281

ABSTRACT

OBJECTIVE: The supramammillary nucleus (SuM) is nestled between the lateral hypothalamus (LH) and the ventral tegmental area (VTA). This neuroanatomical position is consistent with a potential role of this nucleus to regulate ingestive and motivated behavior. Here neuroanatomical, molecular, and behavior approaches are utilized to determine whether SuM contributes to ingestive and food-motivated behavior control. METHODS: Through the application of anterograde and retrograde neural tract tracing with novel designer viral vectors, the current findings show that SuM neurons densely innervate the LH in a sex dimorphic fashion. Glucagon-like peptide-1 (GLP-1) is a clinically targeted neuro-intestinal hormone with a well-established role in regulating energy balance and reward behaviors. Here we determine that GLP-1 receptors (GLP-1R) are expressed throughout the SuM of both sexes, and also directly on SuM LH-projecting neurons and investigate the role of SuM GLP-1R in the regulation of ingestive and motivated behavior in male and female rats. RESULTS: SuM microinjections of the GLP-1 analogue, exendin-4, reduced ad libitum intake of chow, fat, or sugar solution in both male and female rats, while food-motivated behaviors, measured using the sucrose motivated operant conditioning test, was only reduced in male rats. These data contrasted with the results obtained from a neighboring structure well known for its role in motivation and reward, the VTA, where females displayed a more potent response to GLP-1R activation by exendin-4. In order to determine the physiological role of SuM GLP-1R signaling regulation of energy balance, we utilized an adeno-associated viral vector to site-specifically deliver shRNA for the GLP-1R to the SuM. Surprisingly, and in contrast to previous results for the two SuM neighboring sites, LH and VTA, SuM GLP-1R knockdown increased food seeking and adiposity in obese male rats without altering food intake, body weight or food motivation in lean or obese, female or male rats. CONCLUSION: Taken together, these results indicate that SuM potently contributes to ingestive and motivated behavior control; an effect contingent on sex, diet/homeostatic energy balance state and behavior of interest. These data also extend the map of brain sites directly responsive to GLP-1 agonists, and highlight key differences in the role that GLP-1R play in interconnected and neighboring nuclei.


Subject(s)
Basal Ganglia/metabolism , Eating , Glucagon-Like Peptide 1/metabolism , Hypothalamus/metabolism , Motivation , Animals , Basal Ganglia/cytology , Basal Ganglia/physiology , Conditioning, Operant , Energy Metabolism , Female , Glucagon-Like Peptide-1 Receptor/genetics , Glucagon-Like Peptide-1 Receptor/metabolism , Hypothalamus/cytology , Hypothalamus/physiology , Male , Neural Pathways/cytology , Neural Pathways/metabolism , Neural Pathways/physiology , Rats , Rats, Sprague-Dawley , Sex Factors
11.
J Biosci ; 43(5): 921-929, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30541952

ABSTRACT

5rolGLP-HV is a promising dual-function peptide for the treatment of diabetes and thrombosis simultaneously. For investigating the therapeutic mechanism of 5rolGLP-HV for type 2 diabetes mellitus (T2DM), STZ-induced diabetic mice were established and treated with 5rolGLP-HV. The results showed that daily water and food intake, blood glucose, serum and pancreatic insulin levels significantly decreased after 5rolGLP-HV treatment with various oral concentrations, and 16 mg/kg was the optimal dose for controlling diabetes. 5rolGLP-HV treatment decreased the MDA levels and the T-SOD activity in serum and pancreatic of diabetic mice (but not up to significant difference), and significantly increased the expression of signal pathways related genes of rolGLP-1, also the density of insulin expression and the numbers of apoptosis cells in islets of diabetic mice were significantly decreased in comparison to the negative diabetic mice. These effects above may be clarified the hypoglycemic mechanisms of 5rolGLP-HV, and 5rolGLP-HV may be as a potential drug for diabetes in future.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Glucagon-Like Peptide 1/pharmacology , Hypoglycemic Agents/pharmacology , Insulin/blood , Recombinant Proteins/pharmacology , Animals , Blood Glucose/drug effects , Blood Glucose/metabolism , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Drinking/drug effects , Drug Evaluation, Preclinical , Eating/drug effects , Gene Expression Regulation , Glucagon-Like Peptide 1/biosynthesis , Glucagon-Like Peptide-1 Receptor/genetics , Glucagon-Like Peptide-1 Receptor/metabolism , Hirudins/chemistry , Hypoglycemic Agents/metabolism , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/metabolism , Male , Malondialdehyde/antagonists & inhibitors , Malondialdehyde/metabolism , Mice , Mice, Inbred C57BL , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Recombinant Proteins/biosynthesis , Streptozocin , Superoxide Dismutase/metabolism
12.
Nutrients ; 10(10)2018 Sep 21.
Article in English | MEDLINE | ID: mdl-30241419

ABSTRACT

Obesity is one of the major public health issues, and its prevalence is steadily increasing all the world over. The endocannabinoid system (ECS) has been shown to be involved in the intake of palatable food via activation of cannabinoid 1 receptor (CB1R). However, the involvement of lingual CB1R in the orosensory perception of dietary fatty acids has never been investigated. In the present study, behavioral tests on CB1R-/- and wild type (WT) mice showed that the invalidation of Cb1r gene was associated with low preference for solutions containing rapeseed oil or a long-chain fatty acid (LCFA), such as linoleic acid (LA). Administration of rimonabant, a CB1R inverse agonist, in mice also brought about a low preference for dietary fat. No difference in CD36 and GPR120 protein expressions were observed in taste bud cells (TBC) from WT and CB1R-/- mice. However, LCFA induced a higher increase in [Ca2+]i in TBC from WT mice than that in TBC from CB1R-/- mice. TBC from CB1R-/- mice also exhibited decreased Proglucagon and Glp-1r mRNA and a low GLP-1 basal level. We report that CB1R is involved in fat taste perception via calcium signaling and GLP-1 secretion.


Subject(s)
Fatty Acids , Food Preferences , Obesity/genetics , Receptor, Cannabinoid, CB1/genetics , Taste Buds/metabolism , Taste Perception/genetics , Taste/genetics , Animals , CD36 Antigens/genetics , CD36 Antigens/metabolism , Calcium Signaling/genetics , Cannabinoid Receptor Antagonists/pharmacology , Dietary Fats , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor/genetics , Glucagon-Like Peptide-1 Receptor/metabolism , Linoleic Acid , Male , Mice, Knockout , Obesity/etiology , Proglucagon/genetics , Proglucagon/metabolism , RNA, Messenger/metabolism , Rapeseed Oil , Receptor, Cannabinoid, CB1/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Rimonabant/pharmacology
13.
Diabetes ; 67(8): 1538-1548, 2018 08.
Article in English | MEDLINE | ID: mdl-29776968

ABSTRACT

Glucagon-like peptide 1 receptor (GLP-1R) agonists are U.S. Food and Drug Administration-approved weight loss drugs. Despite their widespread use, the sites of action through which GLP-1R agonists (GLP1RAs) affect appetite and body weight are still not fully understood. We determined whether GLP-1Rs in either GABAergic or glutamatergic neurons are necessary for the short- and long-term effects of the GLP1RA liraglutide on food intake, visceral illness, body weight, and neural network activation. We found that mice lacking GLP-1Rs in vGAT-expressing GABAergic neurons responded identically to controls in all parameters measured, whereas deletion of GLP-1Rs in vGlut2-expressing glutamatergic neurons eliminated liraglutide-induced weight loss and visceral illness and severely attenuated its effects on feeding. Concomitantly, deletion of GLP-1Rs from glutamatergic neurons completely abolished the neural network activation observed after liraglutide administration. We conclude that liraglutide activates a dispersed but discrete neural network to mediate its physiological effects and that these effects require GLP-1R expression on glutamatergic but not GABAergic neurons.


Subject(s)
Appetite Depressants/therapeutic use , Glucagon-Like Peptide-1 Receptor/agonists , Hypoglycemic Agents/therapeutic use , Hypothalamus/drug effects , Liraglutide/therapeutic use , Neurons/drug effects , Obesity/drug therapy , Animals , Diet, High-Fat/adverse effects , Energy Intake/drug effects , GABAergic Neurons/drug effects , GABAergic Neurons/metabolism , Genes, Reporter/drug effects , Glucagon-Like Peptide-1 Receptor/chemistry , Glucagon-Like Peptide-1 Receptor/genetics , Glucagon-Like Peptide-1 Receptor/metabolism , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hypothalamus/metabolism , Hypothalamus/pathology , Male , Mice, Knockout , Mice, Transgenic , Nerve Net/drug effects , Nerve Net/metabolism , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Neurons/pathology , Obesity/etiology , Obesity/metabolism , Obesity/pathology , Random Allocation , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Vesicular Glutamate Transport Protein 2/chemistry , Vesicular Glutamate Transport Protein 2/genetics , Vesicular Glutamate Transport Protein 2/metabolism , Vesicular Inhibitory Amino Acid Transport Proteins/chemistry , Vesicular Inhibitory Amino Acid Transport Proteins/genetics , Vesicular Inhibitory Amino Acid Transport Proteins/metabolism , Weight Loss/drug effects
14.
Mol Metab ; 11: 33-46, 2018 05.
Article in English | MEDLINE | ID: mdl-29650350

ABSTRACT

OBJECTIVE: Glucagon-like peptide-1 (GLP-1) neurons in the hindbrain densely innervate the dorsomedial hypothalamus (DMH), a nucleus strongly implicated in body weight regulation and the sympathetic control of brown adipose tissue (BAT) thermogenesis. Therefore, DMH GLP-1 receptors (GLP-1R) are well placed to regulate energy balance by controlling sympathetic outflow and BAT function. METHODS: We investigate this possibility in adult male rats by using direct administration of GLP-1 (0.5 ug) into the DMH, knocking down DMH GLP-1R mRNA with viral-mediated RNA interference, and by examining the neurochemical phenotype of GLP-1R expressing cells in the DMH using in situ hybridization. RESULTS: GLP-1 administered into the DMH increased BAT thermogenesis and hepatic triglyceride (TG) mobilization. On the other hand, Glp1r knockdown (KD) in the DMH increased body weight gain and adiposity, with a concomitant reduction in energy expenditure (EE), BAT temperature, and uncoupling protein 1 (UCP1) expression. Moreover, DMH Glp1r KD induced hepatic steatosis, increased plasma TG, and elevated liver specific de-novo lipogenesis, effects that collectively contributed to insulin resistance. Interestingly, DMH Glp1r KD increased neuropeptide Y (NPY) mRNA expression in the DMH. GLP-1R mRNA in the DMH, however, was found in GABAergic not NPY neurons, consistent with a GLP-1R-dependent inhibition of NPY neurons that is mediated by local GABAergic neurons. Finally, DMH Glp1r KD attenuated the anorexigenic effects of the GLP-1R agonist exendin-4, highlighting an important role of DMH GLP-1R signaling in GLP-1-based therapies. CONCLUSIONS: Collectively, our data show that DMH GLP-1R signaling plays a key role for BAT thermogenesis and adiposity.


Subject(s)
Adipose Tissue, Brown/metabolism , Adiposity , Glucagon-Like Peptide-1 Receptor/metabolism , Hypothalamus/metabolism , Thermogenesis , Animals , Exenatide/metabolism , GABAergic Neurons/metabolism , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor/genetics , Insulin Resistance , Lipogenesis , Male , Neuropeptide Y/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction , Uncoupling Protein 1/metabolism
15.
Peptides ; 100: 140-149, 2018 02.
Article in English | MEDLINE | ID: mdl-29412813

ABSTRACT

Pharmacological long lasting Glucagon-like peptide-1 (GLP-1) analogues, such as Exendin-4, have become widely used diabetes therapies. Chronic GLP-1R stimulation has been linked to ß-cell protection and these pro-survival actions of GLP-1 are dependent on the activation of the mammalian target of rapamycin (mTOR) leading to accumulation of Hypoxia inducible factor 1 alpha (HIF-1α). Recent studies from our lab indicate that prolonged GLP-1R stimulation promotes metabolic reprograming of ß-cells towards a highly glycolytic phenotype and activation of the mTOR/HIF-1α pathway was required for this action. We hypothesised that GLP-1 induced metabolic changes depend on the activation of mTOR and HIF-1α, in a cascade that occurs after triggering of a potential Insulin-like growth factor 1 receptor (IGF-1R) or the Insulin receptor (IR) autocrine loops. Loss of function of these receptors, through the use of small interfering RNA, or neutralizing antibodies directed towards their products, was undertaken in conjunction with functional assays. Neither of these strategies mitigated the effect of GLP-1 on glucose uptake, protein expression or bioenergetic flux. Our data indicates that activation of IGF-1R and/or the IR autocrine loops resulting in ß-cell protection and function, involve mechanisms independent to the enhanced metabolic effects resulting from sustained GLP-1R activation.


Subject(s)
Diabetes Mellitus/drug therapy , Glucagon-Like Peptide-1 Receptor/genetics , Receptor, IGF Type 1/genetics , Receptor, Insulin/genetics , Animals , Autocrine Communication/drug effects , Diabetes Mellitus/metabolism , Diabetes Mellitus/pathology , Energy Metabolism/drug effects , Exenatide/administration & dosage , Glucose/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Insulin/genetics , Insulin-Secreting Cells/drug effects , Rats , TOR Serine-Threonine Kinases/genetics
16.
Zhen Ci Yan Jiu ; 42(2): 107-13, 2017 Apr 25.
Article in Chinese | MEDLINE | ID: mdl-29071956

ABSTRACT

OBJECTIVE: To observe the effect of electroacupuncture (EA) on pancreatic glucagon-like peptide-1 receptor (GLP-1 R), pancreatic and duodenal homeobox 1 (PDX-1) protein expression and blood glucose in type 2 diabetes rats, so as to explore the underlying mechanism of EA treatment in improving type 2 diabetes. METHODS: Sprague-Dawley rats were randomly divided into blank control group, model group, "Weiwanxiashu" (EX-B 3) group, "Xinshu" (BL 15) group, and "Shenshu" (BL 23) group, 12 rats in each group. Diabetes model was established by feeding the rat with high fat and high sugar diet combined with intraperitoneal injection of streptozotocin (35 mg/kg). All the EA groups received 2 Hz, 2 mA continuous wave treatment for 20 min everyday, 6 times per week lasting for 4 weeks. Fasting blood glucose was measured by Roche glucometer before and after treatment. Hematoxylin and eosin (HE) staining and Masson staining were used to detect pancreas morphology. GLP-1 R and PDX-1 protein expressions in the pancreas were detected by Western blot. RESULTS: Compared to the blank control group, fasting blood glucose was significantly increased in the model group(P<0.01), accompanied with shrunken islet area, reduced nucleus counts of islet ß cells, and compensatorily enlarged ß cell nucleus. Compared to the model group, EA intervention significantly reduced fasting blood glucose level only in the EX-B 3 group (P<0.05), partly restored pancreas morphology and nucleus counts of islet ß cells in the EX-B 3, BL 15, and BL 23 groups. Compared to the blank control group, GLP-1 R and PDX-1 expressions were decreased in the model group (P<0.01), while EA treatment could obviously increase GLP-1 R expression in the EX-B 3(P<0.01), BL 15 (P<0.01) and BL 23 (P<0.05) groups compared with the model group. The expression of GLP-1 R in the BL 15 group was the highest among the three EA groups (P<0.05,P<0.01), and that in the EX-B 3 group was higher than in the BL 23 group (P<0.05).There were no signifincant differences in the expression of PDX-1 protein among the three EA groups (P>0.05). CONCLUSIONS: EA treatment at EX-B 3 can reduce blood glucose via regulating pancreas function, increasing pancreatic GLP-1 R expression, and partly restoring the morphology of pancreas.


Subject(s)
Acupuncture Points , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/therapy , Electroacupuncture , Glucagon-Like Peptide-1 Receptor/metabolism , Islets of Langerhans/anatomy & histology , Pancrelipase/metabolism , Animals , Diabetes Mellitus, Type 2/genetics , Glucagon/metabolism , Glucagon-Like Peptide-1 Receptor/genetics , Humans , Islets of Langerhans/metabolism , Male , Rats , Rats, Sprague-Dawley
17.
Mol Metab ; 6(7): 748-759, 2017 07.
Article in English | MEDLINE | ID: mdl-28702330

ABSTRACT

OBJECTIVE: Adult obesity risk is influenced by alterations to fetal and neonatal environments. Modifying neonatal gut or neurohormone signaling pathways can have negative metabolic consequences in adulthood. Here we characterize the effect of neonatal activation of glucagon like peptide-1 (GLP-1) receptor (GLP1R) signaling on adult adiposity and metabolism. METHODS: Wild type C57BL/6 mice were injected with 1 nmol/kg Exendin-4 (Ex-4), a GLP1R agonist, for 6 consecutive days after birth. Growth, body composition, serum analysis, energy expenditure, food intake, and brain and fat pad histology and gene expression were assessed at multiple time points through 42 weeks. Similar analyses were conducted in a Glp1r conditional allele crossed with a Sim1Cre deleter strain to produce Sim1Cre;Glp1rloxP/loxP mice and control littermates. RESULTS: Neonatal administration of Ex-4 reduced adult body weight and fat mass, increased energy expenditure, and conferred protection from diet-induced obesity in female mice. This was associated with induction of brown adipose genes and increased noradrenergic fiber density in parametrial white adipose tissue (WAT). We further observed durable alterations in orexigenic and anorexigenic projections to the paraventricular hypothalamic nucleus (PVH). Genetic deletion of Glp1r in the PVH by Sim1-Cre abrogated the impact of neonatal Ex-4 on adult body weight, WAT browning, and hypothalamic architecture. CONCLUSION: These observations suggest that the acute activation of GLP1R in neonates durably alters hypothalamic architecture to limit adult weight gain and adiposity, identifying GLP1R as a therapeutic target for obesity prevention.


Subject(s)
Adiposity , Glucagon-Like Peptide-1 Receptor/agonists , Hypothalamus/growth & development , Animals , Exenatide , Female , Glucagon-Like Peptide-1 Receptor/genetics , Glucagon-Like Peptide-1 Receptor/metabolism , Hypothalamus/cytology , Incretins/pharmacology , Male , Mice , Mice, Inbred C57BL , Neurons/cytology , Neurons/metabolism , Orexins/genetics , Orexins/metabolism , Peptides/pharmacology , Venoms/pharmacology
18.
Diabetes ; 66(2): 372-384, 2017 02.
Article in English | MEDLINE | ID: mdl-27908915

ABSTRACT

Pharmacological activation of the hypothalamic glucagon-like peptide 1 (GLP-1) receptor (GLP-1R) promotes weight loss and improves glucose tolerance. This demonstrates that the hypothalamic GLP-1R is sufficient but does not show whether it is necessary for the effects of exogenous GLP-1R agonists (GLP-1RA) or endogenous GLP-1 on these parameters. To address this, we crossed mice harboring floxed Glp1r alleles to mice expressing Nkx2.1-Cre to knock down Glp1r expression throughout the hypothalamus (GLP-1RKDΔNkx2.1cre). We also generated mice lacking Glp1r expression specifically in two GLP-1RA-responsive hypothalamic feeding nuclei/cell types, the paraventricular nucleus (GLP-1RKDΔSim1cre) and proopiomelanocortin neurons (GLP-1RKDΔPOMCcre). Chow-fed GLP-1RKDΔNkx2.1cre mice exhibited increased food intake and energy expenditure with no net effect on body weight. When fed a high-fat diet, these mice exhibited normal food intake but elevated energy expenditure, yielding reduced weight gain. None of these phenotypes were observed in GLP-1RKDΔSim1cre and GLP-1RKDΔPOMCcre mice. The acute anorectic and glucose tolerance effects of peripherally dosed GLP-1RA exendin-4 and liraglutide were preserved in all mouse lines. Chronic liraglutide treatment reduced body weight in chow-fed GLP-1RKDΔNkx2.1cre mice, but this effect was attenuated with high-fat diet feeding. In sum, classic homeostatic control regions are sufficient but not individually necessary for the effects of GLP-1RA on nutrient homeostasis.


Subject(s)
Eating/genetics , Energy Metabolism/genetics , Glucagon-Like Peptide-1 Receptor/genetics , Glucose/metabolism , Hypothalamus/metabolism , Animals , Body Composition , Diet, High-Fat , Eating/drug effects , Exenatide , Gene Knockdown Techniques , Glucagon-Like Peptide 1/pharmacology , Glucagon-Like Peptide-1 Receptor/metabolism , Glucose Tolerance Test , Homeostasis/genetics , Incretins/pharmacology , Liraglutide/pharmacology , Male , Mice , Neurons/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Peptides/pharmacology , Pro-Opiomelanocortin/metabolism , Venoms/pharmacology , Weight Gain/drug effects , Weight Gain/genetics
19.
J Clin Endocrinol Metab ; 101(5): 2122-9, 2016 05.
Article in English | MEDLINE | ID: mdl-26672638

ABSTRACT

CONTEXT: Glucagon-like peptide-1 (GLP-1) and GLP-1 receptor agonist treatment in type 2 diabetes (T2DM) reduce blood glucose and food intake. It has been suggested that these effects are partly mediated through central GLP-1 receptors (GLP-1Rs). The rodent and nonhuman primate hypothalamus show clear GLP-1R expression. However, a detailed description of GLP-1R expression in the human hypothalamus is lacking, and it is unknown whether this expression is altered in T2DM patients. OBJECTIVE: The objective of the study was to investigate the GLP-1R distribution in the human postmortem hypothalamus and to determine whether hypothalamic GLP-1R expression is altered in T2DM patients. DESIGN: We investigated the distribution of GLP-1R expression throughout the human hypothalamus by means of in situ hybridization. We also performed quantifications of GLP-1R mRNA expression in two hypothalamic nuclei (ie, the paraventricular nucleus [PVN] and infundibular nucleus [IFN]), comparing patients with T2DM and control subjects. RESULTS: We found that GLP-1R mRNA was expressed in a number of hypothalamic nuclei including the PVN and the IFN, both involved in the regulation of energy metabolism. We observed sporadic colocalization of the GLP-1R in the IFN with the orgexigenic neuropeptide Y, agouti-related peptide, or proopiomelanocortin transcripts. Comparison of GLP-1R mRNA in the PVN and IFN between T2DM patients and control subjects revealed a decreased expression in T2DM patients. CONCLUSIONS: Our studies show that GLP-1R is widely expressed throughout the human hypothalamus. The decreased expression of GLP-1R in the PVN and IFN of T2DM patients may be related to the dysregulation of feeding behavior and glucose homeostasis in T2DM.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Glucagon-Like Peptide-1 Receptor/metabolism , Hypothalamus/metabolism , Aged , Aged, 80 and over , Agouti-Related Protein/genetics , Agouti-Related Protein/metabolism , Arcuate Nucleus of Hypothalamus/metabolism , Diabetes Mellitus, Type 2/genetics , Female , Glucagon-Like Peptide-1 Receptor/genetics , Humans , Male , Middle Aged , Neuropeptide Y/genetics , Neuropeptide Y/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Pro-Opiomelanocortin/genetics , Pro-Opiomelanocortin/metabolism
20.
Nat Commun ; 6: 8918, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26621478

ABSTRACT

Glucagon-like peptide-1 (GLP-1) receptor (GLP-1R) agonists have emerged as treatment options for type 2 diabetes mellitus (T2DM). GLP-1R signals through G-protein-dependent, and G-protein-independent pathways by engaging the scaffold protein ß-arrestin; preferential signalling of ligands through one or the other of these branches is known as 'ligand bias'. Here we report the discovery of the potent and selective GLP-1R G-protein-biased agonist, P5. We identified P5 in a high-throughput autocrine-based screening of large combinatorial peptide libraries, and show that P5 promotes G-protein signalling comparable to GLP-1 and Exendin-4, but exhibited a significantly reduced ß-arrestin response. Preclinical studies using different mouse models of T2DM demonstrate that P5 is a weak insulin secretagogue. Nevertheless, chronic treatment of diabetic mice with P5 increased adipogenesis, reduced adipose tissue inflammation as well as hepatic steatosis and was more effective at correcting hyperglycaemia and lowering haemoglobin A1c levels than Exendin-4, suggesting that GLP-1R G-protein-biased agonists may provide a novel therapeutic approach to T2DM.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Glucagon-Like Peptide-1 Receptor/antagonists & inhibitors , Hypoglycemic Agents/administration & dosage , Peptides/administration & dosage , Adipogenesis/drug effects , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/physiopathology , Drug Evaluation, Preclinical , Glucagon-Like Peptide-1 Receptor/genetics , Glucagon-Like Peptide-1 Receptor/metabolism , Humans , Insulin/metabolism , Male , Mice , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL