Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Mol Metab ; 6(6): 503-511, 2017 06.
Article in English | MEDLINE | ID: mdl-28580281

ABSTRACT

OBJECTIVE: Glucagon-like peptides (GLPs) are secreted from enteroendocrine cells in response to nutrients and bile acids and control metabolism via actions on structurally-related yet distinct G protein coupled receptors. GLP-1 regulates gut motility, appetite, islet function, and glucose homeostasis, whereas GLP-2 enhances intestinal nutrient absorption. GLP-1R agonists are used to treat diabetes and obesity, and a GLP-2R agonist is approved to treat short bowel syndrome. Unexpectedly, reports of gallbladder disease have been associated with the use of both GLP-1R and GLP-2R agonists and after bariatric surgery, although the mechanisms remain unknown. METHODS: We investigated whether GLP-1 or GLP-2 acutely controls gallbladder (GB) volume and whether GLP-2 regulates GB muscle activity in mice. The expression of Tgr5, Glp2r, and Glp1r was assessed in mouse GB, and the effects of GLP-2 on hepatic bile acid (BA) flow, intestinal and liver BA uptake, and GB gene expression were determined. GLP-2 regulation of GB volume was assessed in wildtype, Glp2r-/- and Tgr5-/- mice. The effect of GLP-2 on GB smooth muscle (GBSM) calcium transients was characterized ex vivo. RESULTS: Acute administration of the GLP-1R agonist exendin-4 lowered glucose but had no effect on GB volume in mice. In contrast, GLP-2 rapidly enhanced GB filling in a dose-dependent manner, actions maintained in the presence of cholecystokinin, and mediated through the canonical GLP-2R. GLP-2 also rapidly induced immediate early gene expression in GB, consistent with detection of the endogenous Glp2r in GB RNA. The ability of GLP-2 to increase GB volume was not abrogated by systemic administration of hexamethonium, propranolol, a vasoactive peptide receptor antagonist or N-Nitroarginine methyl ester, and was maintained in Tgr5-/- mice. In contrast, lithocholic acid, a Tgr5 agonist, increased GB filling in Glp2r-/- but not in Tgr5-/- mice. GLP-2 had no effect on ileal uptake or hepatic clearance of taurocholic acid or on hepatic bile flow, yet reduced the frequency of spontaneous calcium transients in murine GBSM ex vivo, in a tetrodotoxin-sensitive manner. CONCLUSIONS: Our data extend endocrine concepts of regulation of GB filling beyond FXR-FGF15/19 and the direct effects of BA via Tgr5, to encompass a novel BA-Tgr5-L cell GLP-2 axis providing nutrient-mediated feedback from BA to terminate meal-related GB contraction. These findings have implications for conditions characterized by elevated circulating levels of GLP-2 such as after bariatric surgery and the development and use of agents that promote Tgr5 activation, L cell secretion, or GLP-2R agonism for the treatment of metabolic disease.


Subject(s)
Gallbladder/drug effects , Gastrointestinal Agents/pharmacology , Glucagon-Like Peptide-2 Receptor/metabolism , Peptides/pharmacology , Receptors, G-Protein-Coupled/metabolism , Animals , Gallbladder/metabolism , Gallbladder/physiology , Humans , Male , Mice , Mice, Inbred C57BL , Muscle Contraction , Receptors, G-Protein-Coupled/genetics
2.
Diabetes ; 66(6): 1626-1635, 2017 06.
Article in English | MEDLINE | ID: mdl-28254842

ABSTRACT

GPR119 was originally identified as an orphan ß-cell receptor; however, subsequent studies demonstrated that GPR119 also regulates ß-cell function indirectly through incretin hormone secretion. We assessed the importance of GPR119 for ß-cell function in Gpr119-/- mice and in newly generated Gpr119ßcell-/- mice. Gpr119-/- mice displayed normal body weight and glucose tolerance on a regular chow (RC) diet. After high-fat feeding, Gpr119-/- mice exhibited reduced fat mass, decreased levels of circulating adipokines, improved insulin sensitivity, and better glucose tolerance. Unexpectedly, oral and intraperitoneal glucose tolerance and the insulin response to glycemic challenge were not perturbed in Gpr119ßcell-/- mice on RC and high-fat diets. Moreover, islets from Gpr119-/- and Gpr119ßcell-/- mice exhibited normal insulin responses to glucose and ß-cell secretagogues. Furthermore, the selective GPR119 agonist AR231453 failed to directly enhance insulin secretion from perifused islets. In contrast, AR231453 increased plasma glucagon-like peptide 1 (GLP-1) and insulin levels and improved glucose tolerance in wild-type and Gpr119ßcell-/- mice. These findings demonstrate that ß-cell GPR119 expression is dispensable for the physiological control of insulin secretion and the pharmacological response to GPR119 agonism, findings that may inform the lack of robust efficacy in clinical programs assessing GPR119 agonists for the therapy of type 2 diabetes.


Subject(s)
Blood Glucose/metabolism , Diabetes Mellitus, Experimental/metabolism , Glucagon-Like Peptide 1/metabolism , Insulin Resistance , Insulin-Secreting Cells/metabolism , Receptors, G-Protein-Coupled/genetics , Adipokines/metabolism , Animals , Apoptosis , Diet, High-Fat , Gene Expression Profiling , Glucagon/metabolism , Glucagon-Like Peptide 1/drug effects , Glucose/metabolism , Glucose Tolerance Test , Incretins/metabolism , Insulin/metabolism , Insulin Secretion , Male , Mice, Knockout , Oxadiazoles/pharmacology , Pyrimidines/pharmacology , Receptors, G-Protein-Coupled/agonists
3.
Diabetes ; 62(1): 283-90, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22933106

ABSTRACT

Glucagon-like peptide-1 (GLP-1) secretion is classically regulated by ingested nutrients. To identify novel molecular targets controlling incretin secretion, we analyzed enteroendocrine cell pathways important for hormone biosynthesis and secretion. We demonstrate that progesterone increases GLP-1 secretion and extracellular signal-related kinase 1/2 (ERK1/2) phosphorylation in enteroendocrine GLUTag cells via mechanisms sensitive to the mitogen-activated protein kinase inhibitor U0126. The stimulatory effects of progesterone (P4) or the synthetic progestin R5020 on ERK1/2 phosphorylation were independent of the classical progesterone receptor antagonist RU486. Furthermore, a cell-impermeable BSA-progesterone conjugate rapidly increased ERK1/2 phosphorylation and GLP-1 secretion. Knockdown of the membrane progesterone receptors Paqr5 or Paqr7 in GLUTag cells eliminated the stimulatory effect of R5020 and progesterone on GLP-1 secretion. Enteral progesterone administration increased plasma levels of GLP-1, glucose-dependent insulinotropic polypeptide (GIP), and insulin, and improved oral glucose tolerance in an RU486-insensitve manner in mice: however, systemic progesterone exposure did not improve glucose homeostasis. Unexpectedly, the glucoregulatory actions of enteral progesterone did not require classical incretin receptor signaling and were preserved in Glp1r(-/-) and Glp1r(-/-):Gipr(-/-) mice. Intestine-restricted activation of membrane progesterone receptors may represent a novel approach for stimulation of incretin hormone secretion and control of glucose homeostasis.


Subject(s)
Enteroendocrine Cells/physiology , Glucose/metabolism , Incretins/metabolism , Receptors, Progesterone/physiology , Animals , Extracellular Signal-Regulated MAP Kinases/metabolism , Gastric Inhibitory Polypeptide/blood , Glucagon/biosynthesis , Glucagon-Like Peptide 1/metabolism , Homeostasis , Male , Mice , Mice, Inbred C57BL , Phosphorylation , Promegestone/pharmacology
4.
Endocrinology ; 152(2): 374-83, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21068156

ABSTRACT

G protein-coupled receptor 119 (GPR119) was originally identified as a ß-cell receptor. However, GPR119 activation also promotes incretin secretion and enhances peptide YY action. We examined whether GPR119-dependent control of glucose homeostasis requires preservation of peptidergic pathways in vivo. Insulin secretion was assessed directly in islets, and glucoregulation was examined in wild-type (WT), single incretin receptor (IR) and dual IR knockout (DIRKO) mice. Experimental endpoints included plasma glucose, insulin, glucagon, glucagon-like peptide-1 (GLP-1), glucose-dependent insulinotropic peptide (GIP), and peptide YY. Gastric emptying was assessed in WT, Glp1r-/-, DIRKO, Glp2r-/-, and GPR119-/- mice treated with the GPR119 agonist AR231453. AR231453 stimulated insulin secretion from WT and DIRKO islets in a glucose-dependent manner, improved glucose homeostasis, and augmented plasma levels of GLP-1, GIP, and insulin in WT and Gipr-/- mice. In contrast, although AR231453 increased levels of GLP-1, GIP, and insulin, it failed to lower glucose in Glp1r-/- and DIRKO mice. Furthermore, AR231453 did not improve ip glucose tolerance and had no effect on insulin action in WT and DIRKO mice. Acute GPR119 activation with AR231453 inhibited gastric emptying in Glp1r-/-, DIRKO, Glp2r-/-, and in WT mice independent of the Y2 receptor (Y2R); however, AR231453 did not control gastric emptying in GPR119-/- mice. Our findings demonstrate that GPR119 activation directly stimulates insulin secretion from islets in vitro, yet requires intact IR signaling and enteral glucose exposure for optimal control of glucose tolerance in vivo. In contrast, AR231453 inhibits gastric emptying independent of incretin, Y2R, or Glp2 receptors through GPR119-dependent pathways. Hence, GPR119 engages multiple complementary pathways for control of glucose homeostasis.


Subject(s)
Blood Glucose/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Gastrointestinal Hormone/metabolism , Animals , Blood Glucose/genetics , Gastric Emptying/drug effects , Gastric Emptying/genetics , Gastric Inhibitory Polypeptide/blood , Glucagon/blood , Glucagon-Like Peptide 1/blood , Glucose Tolerance Test , Insulin/blood , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxadiazoles/pharmacology , Pyrimidines/pharmacology , Receptors, G-Protein-Coupled/genetics , Receptors, Gastrointestinal Hormone/genetics
5.
Diabetes ; 57(2): 288-97, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17977951

ABSTRACT

OBJECTIVE: The incretins glucagon-like peptide 1 and glucose-dependent insulinotropic polypeptide have been postulated to play a role in regulating insulin action, although the mechanisms behind this relationship remain obscure. We used the hyperinsulinemic-euglycemic clamp to determine sites where insulin action may be modulated in double incretin receptor knockout (DIRKO) mice, which lack endogenous incretin action. RESEARCH DESIGN AND METHODS: DIRKO and wild-type mice were fed regular chow or high-fat diet for 4 months. Clamps were performed on 5-h-fasted, conscious, unrestrained mice using an arterial catheter for sampling. RESULTS: Compared with wild-type mice, chow and high fat-fed DIRKO mice exhibited decreased fat and muscle mass associated with increased energy expenditure and ambulatory activity. Clamp rates of glucose infusion (GIR), endogenous glucose production (endoR(a)), and disappearance (R(d)) were not different in chow-fed wild-type and DIRKO mice, although insulin levels were lower in DIRKO mice. Liver Akt expression was decreased but Akt activation was increased in chow-fed DIRKO compared with wild-type mice. High-fat feeding resulted in fasting hyperinsulinemia and hyperglycemia in wild-type but not in DIRKO mice. GIR, suppression of endoR(a), and stimulation of R(d) were inhibited in high fat-fed wild-type mice but not in DIRKO mice. High-fat feeding resulted in impaired tissue glucose uptake (R(g)) in skeletal muscle of wild-type mice but not of DIRKO mice. Liver and muscle Akt activation was enhanced in high fat-fed DIRKO compared with wild-type mice. CONCLUSIONS: In summary, DIRKO mice exhibit enhanced insulin action compared with wild-type mice when fed a regular chow diet and are protected from high-fat diet-induced obesity and insulin resistance.


Subject(s)
Insulin/pharmacology , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/genetics , Receptors, Gastrointestinal Hormone/deficiency , Receptors, Gastrointestinal Hormone/genetics , Receptors, Glucagon/deficiency , Receptors, Glucagon/genetics , Adipose Tissue/anatomy & histology , Animals , Crosses, Genetic , Dietary Fats , Energy Metabolism , Female , Glucagon-Like Peptide-1 Receptor , Glucose Clamp Technique , Hyperinsulinism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/anatomy & histology
6.
Diabetes ; 56(12): 3006-13, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17717280

ABSTRACT

OBJECTIVE: Dipeptidyl peptidase-4 (DPP4) inhibitors lower blood glucose in diabetic subjects; however, the mechanism of action through which these agents improve glucose homeostasis remains incompletely understood. Although glucagon-like peptide (GLP)-1 and glucose-dependent insulinotropic polypeptide (GIP) represent important targets for DPP4 activity, whether additional substrates are important for the glucose-lowering actions of DPP4 inhibitors remains uncertain. RESEARCH DESIGN AND METHODS: We examined the efficacy of continuous vildagliptin administration in wild-type (WT) and dual incretin receptor knockout (DIRKO) mice after 8 weeks of a high-fat diet. RESULTS: Vildagliptin had no significant effect on food intake, energy expenditure, body composition, body weight gain, or insulin sensitivity in WT or DIRKO mice. However, glycemic excursion after oral glucose challenge was significantly reduced in WT but not in DIRKO mice after vildagliptin treatment. Moreover, vildagliptin increased levels of glucose-stimulated plasma insulin and reduced levels of cholesterol and triglycerides in WT but not in DIRKO mice. Vildagliptin treatment reduced the hepatic expression of genes important for cholesterol synthesis and fatty acid oxidation, including phospho-mevalonate kinase (Mvk), acyl-coenzyme dehydrogenase medium chain (Acadm), mevalonate (diphospho)decarboxylase (Mvd), and Acyl-CoA synthetase (Acsl1), in WT but not in DIRKO mice. However, vildagliptin also reduced levels of hepatic mRNA transcripts for farnesyl di-phosphate transferase (Fdft1), acetyl coenzyme A acyltransferase 1 (Acaa1), and carnitine palmitoyl transferase 1 (Cpt 1) in DIRKO mice. No direct effect of GLP-1 receptor agonists was detected on cholesterol or triglyceride synthesis and secretion in WT hepatocytes. CONCLUSIONS: These findings illustrate that although GLP-1 and GIP receptors represent the dominant molecular mechanisms for transducing the glucoregulatory actions of DPP4 inhibitors, prolonged DPP4 inhibition modulates the expression of genes important for lipid metabolism independent of incretin receptor action in vivo.


Subject(s)
Adamantane/analogs & derivatives , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Hypoglycemic Agents/therapeutic use , Incretins/physiology , Nitriles/therapeutic use , Pyrrolidines/therapeutic use , Receptors, Gastrointestinal Hormone/physiology , Receptors, Glucagon/physiology , Receptors, Peptide/physiology , Adamantane/therapeutic use , Animals , Blood Glucose/drug effects , Blood Glucose/metabolism , Cell Division/drug effects , Glucagon-Like Peptide-1 Receptor , Glucose Tolerance Test , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/physiology , Mice , Receptors, Gastrointestinal Hormone/drug effects , Receptors, Glucagon/drug effects , Receptors, Peptide/deficiency , Receptors, Peptide/genetics , Vildagliptin
7.
J Clin Invest ; 117(1): 143-52, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17187081

ABSTRACT

The incretin hormones glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) control glucose homeostasis through well-defined actions on the islet beta cell via stimulation of insulin secretion and preservation and expansion of beta cell mass. We examined the importance of endogenous incretin receptors for control of glucose homeostasis through analysis of Glp1r(-/-), Gipr(-/-), and double incretin receptor knockout (DIRKO) mice fed a high-fat (HF) diet. DIRKO mice failed to upregulate levels of plasma insulin, pancreatic insulin mRNA transcripts, and insulin content following several months of HF feeding. Both single incretin receptor knockout and DIRKO mice exhibited resistance to diet-induced obesity, preservation of insulin sensitivity, and increased energy expenditure associated with increased locomotor activity. Moreover, plasma levels of plasminogen activator inhibitor-1 and resistin failed to increase significantly in DIRKO mice after HF feeding, and the GIP receptor agonist [D-Ala(2)]GIP, but not the GLP-1 receptor agonist exendin-4, increased the levels of plasma resistin in studies of both acute and chronic administration. These findings extend our understanding of how endogenous incretin circuits regulate glucose homeostasis independent of the beta cell via control of adipokine secretion and energy expenditure.


Subject(s)
Body Weight/physiology , Energy Metabolism/physiology , Receptors, Glucagon/physiology , Animals , Glucagon-Like Peptide-1 Receptor , Homeostasis , Insulin/genetics , Islets of Langerhans/physiology , Mice , Mice, Knockout , RNA, Messenger/genetics , Receptors, Glucagon/deficiency , Receptors, Glucagon/genetics , Transcription, Genetic
8.
Endocrinology ; 146(1): 441-9, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15471960

ABSTRACT

Pdx-1 plays a key role in the development of the pancreas and the control of islet gene transcription and has also been proposed as a dominant regulator of the alpha- vs. beta-cell phenotype via extinction of proglucagon expression. To ascertain the relationship between Pdx-1 and proglucagon gene expression, we examined the effect of enhanced pdx-1 expression on proglucagon gene expression in murine islet alphaTC-1 and GLUTag enteroendocrine cells. Although adenoviral transduction increased the levels of pdx-1 mRNA transcripts and nuclear Pdx-1 protein, overexpression of pdx-1 did not repress endogenous proglucagon gene expression in alphaTC-1 or GLUTag cells or murine islets. Immunohistochemical analysis of cells transduced with Ad-pdx-1 demonstrated multiple individual islet or enteroendocrine cells exhibiting both nuclear Pdx-1 and cytoplasmic glucagon-like peptide-1 immunopositivity. The failure of pdx-1 to inhibit endogenous proglucagon gene expression was not attributable to defects in Pdx-1 nuclear translocation or DNA binding as demonstrated using Western blotting and EMSA analyses. Furthermore, Ad-pdx-1 transduction did not repress proglucagon promoter activity in alphaTC-1 or GLUTag cells. Taken together, these findings demonstrate that pdx-1 alone is not sufficient for specification of the hormonal phenotype or extinction of proglucagon gene expression in islet or enteroendocrine cells.


Subject(s)
Endocrine Glands/metabolism , Glucagon/antagonists & inhibitors , Glucagon/genetics , Homeodomain Proteins/physiology , Intestinal Mucosa/metabolism , Islets of Langerhans/metabolism , Protein Precursors/antagonists & inhibitors , Protein Precursors/genetics , Trans-Activators/physiology , Transcription, Genetic/physiology , Animals , Cell Line , Endocrine Glands/cytology , Glucagon/metabolism , Glucagon-Like Peptide 1 , Homeodomain Proteins/metabolism , Intestines/cytology , Islets of Langerhans/cytology , Mice , Peptide Fragments/metabolism , Proglucagon , Protein Precursors/metabolism , Trans-Activators/metabolism , Transduction, Genetic
9.
Endocrinology ; 144(5): 2025-33, 2003 May.
Article in English | MEDLINE | ID: mdl-12697711

ABSTRACT

Despite interest in understanding glucagon-like peptide-1 (GLP-1) production, the factors important for GLP-1 biosynthesis remain poorly understood. We examined control of human proglucagon gene expression in NCI-H716 cells, a cell line that secretes GLP-1 in a regulated manner. Insulin, phorbol myristate acetate, or forskolin, known regulators of rodent proglucagon gene expression, had no effect, whereas sodium butyrate decreased levels of NCI-H716 proglucagon mRNA transcripts. The inhibitory effect of sodium butyrate was mimicked by trichostatin A but was not detected with sodium acetate or isobutyrate. The actions of butyrate were not diminished by the ERK1/2 inhibitor PD98059, p38 inhibitor SB203580, or soluble guanylate cyclase inhibitor LY83583 or following treatment of cells with KT5823, a selective inhibitor of cGMP-dependent protein kinase. NCI-H716 cells expressed multiple proglucagon gene transcription factors including isl-1, pax-6, pax-2, cdx-2/3, pax-4, hepatocyte nuclear factor (HNF)-3 alpha, HNF-3beta, HNF-3 gamma, and Nkx2.2. Nevertheless, the butyrate-dependent inhibition of proglucagon gene expression was not associated with coordinate changes in transcription factor expression and both the human and rat transfected proglucagon promoters were transcriptionally inactive in NCI-H716 cells. Hence, NCI-H716 cells may not be a physiologically optimal model for studies of human enteroendocrine proglucagon gene transcription.


Subject(s)
Adenocarcinoma/genetics , Cecal Neoplasms/genetics , Gene Expression Regulation/physiology , Glucagon/genetics , Intestines/physiopathology , Protein Precursors/genetics , Adenocarcinoma/metabolism , Animals , Butyrates/pharmacology , Cecal Neoplasms/metabolism , Gene Expression/drug effects , Glucagon/metabolism , Glucagon-Like Peptide 1 , Homeobox Protein Nkx-2.2 , Homeodomain Proteins , Humans , Nuclear Proteins , Peptide Fragments/metabolism , Proglucagon , Protein Precursors/metabolism , Rats , Transcription Factors/metabolism , Tumor Cells, Cultured
10.
J Biol Chem ; 277(48): 46544-51, 2002 Nov 29.
Article in English | MEDLINE | ID: mdl-12297495

ABSTRACT

A mammalian basic helix-loop-helix protein known variably as Stra13, Sharp2, and Dec1 has been implicated in cell activation, proliferation, and differentiation. Indeed, Stra13 null mice develop age-induced autoimmunity as a result of impaired T-lymphocyte activation, leading ultimately to the accumulation of autoreactive T-cells and B-cells. Stra13 is expressed in embryonic as well as adult tissues derived from neuroectoderm, mesoderm, and endoderm and has been associated with response to hypoxia, suggesting a complex role for this protein and the highly related Sharp1/Dec2 protein in homeostatic regulation. Whereas Stra13 is known to regulate many important cellular functions and is known to cross-regulate biological responses to other basic helix-loop-helix containing transcription factors, including c-Myc and USF, it is unclear if this protein binds directly to DNA. Indeed, the basic domain of Stra13 contains a proline residue at an unprecedented position. Herein, we have determined that Stra13 binds with high affinity to CACGTG class B E-box elements as a homodimer with preference for elements preceded by T and/or followed by A residues. In addition, transient transfection experiments reveal that Stra13 represses transcription when bound to these and related sites. Our data suggest that Stra13 regulates cellular functions through antagonism of E-box activator proteins and also through active repression from E-box elements.


Subject(s)
Homeodomain Proteins/physiology , Repressor Proteins/physiology , Animals , Base Sequence , Basic Helix-Loop-Helix Transcription Factors , COS Cells , DNA Probes , Dimerization , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mice , Plasmids , Protein Binding , Repressor Proteins/genetics , Repressor Proteins/metabolism
11.
Mol Endocrinol ; 16(10): 2349-59, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12351699

ABSTRACT

Tissue-specific proglucagon gene transcription is achieved through combinations of transcription factors expressed in pancreatic A cells and enteroendocrine L cells of the small and large intestine. Cell transfection and electrophoretic mobility shift assay experiments previously identified Pax-2 as a regulator of islet proglucagon gene expression. We examined whether Pax-2 regulates gut proglucagon gene expression using enteroendocrine cell lines and Pax2(1NEU) mutant mice. Immunoreactive Pax-2 was detected in STC-1 enteroendocrine cells, and Pax-2 activated proglucagon promoter activity in transfected baby hamster kidney and GLUTag cells. Pax-2 antisera diminished the formation of a Pax-2-G3 complex in electrophoretic mobility shift assay studies using nuclear extracts from islet and enteroendocrine cell lines. Surprisingly, Pax-2 mRNA transcripts were not detected by RT-PCR in RNA isolated from adult rat pancreas, rat islets, embryonic d 19 or adult murine pancreas and gastrointestinal tract. Furthermore, embryonic d 19 or neonatal d 1 Pax2(1NEU) mice exhibited normal islet A cells and gut endocrine L cells, and no decrement in pancreatic or intestinal glucagon gene expression. These findings demonstrate that Pax-2 is not essential for the developmental formation of islet A or gut L cells and does not play a role in the physiological control of proglucagon gene expression in vivo.


Subject(s)
DNA-Binding Proteins/metabolism , Glucagon/genetics , Intestines/cytology , Pancreas/cytology , Promoter Regions, Genetic , Protein Precursors/genetics , Transcription Factors/metabolism , Animals , Animals, Newborn , Cell Lineage , Cells, Cultured , Cricetinae , DNA-Binding Proteins/genetics , Enteroendocrine Cells/metabolism , Gene Expression Regulation , Glucagon/metabolism , Homozygote , Intestines/physiology , Islets of Langerhans/metabolism , Kidney/cytology , Kidney/metabolism , Mice , Mice, Transgenic , PAX2 Transcription Factor , Pancreas/embryology , Pancreas/physiology , Proglucagon , Protein Precursors/metabolism , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...