Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Mol Genet Metab ; 125(1-2): 53-58, 2018 09.
Article in English | MEDLINE | ID: mdl-30064964

ABSTRACT

Mucopolysaccharidosis type II (MPS II or Hunter syndrome) is a lysosomal storage disorder caused by a deficiency of iduronate-2-sulfatase (IDS), an enzyme that catabolizes glycosaminoglycans (GAGs) including heparan sulfate (HS) and dermatan sulfate (DS). GAG accumulation leads to severe neurological and somatic impairments. At present, the most common treatment for MPS II is intravenous enzyme replacement therapy; however, the inability of recombinant IDS to cross the blood-brain barrier (BBB) restricts therapeutic efficacy for neurological manifestations. We recently developed a BBB-penetrating IDS fusion protein, JR-141, and demonstrated its ability to reduce GAG accumulation in the brain of human transferrin receptor knock-in and Ids knock-out mice (TFRC-KI/Ids-KO), an animal model of MPS II, following intravenous administration. Given the impossibility of measuring GAG accumulation in the brains of human patients with MPS II, we hypothesized that GAG content in the cerebrospinal fluid (CSF) might serve as an indicator of brain GAG burden. To test this hypothesis, we optimized a high-sensitivity method for quantifying HS and DS in low-volume samples by combining acidic methanolysis and liquid chromatography-tandem mass spectrometry (LC/MS/MS). We employed this method to quantify HS and DS in samples from TFRC-KI/Ids-KO mice and revealed that HS but not DS accumulated in the central nerve system (CNS). Moreover, concentrations of HS in CSF correlated with those in brain. Finally, intravenous treatment with JR-141 reduced levels of HS in the CSF and brain in TFRC-KI/Ids-KO mice. These results suggest that CSF HS content may be a useful biomarker for evaluating the brain GAG accumulation and the therapeutic efficacy of drugs in patients with MPS II.


Subject(s)
Biomarkers/cerebrospinal fluid , Heparitin Sulfate/cerebrospinal fluid , Mucopolysaccharidosis II/cerebrospinal fluid , Nervous System Diseases/cerebrospinal fluid , Animals , Blood-Brain Barrier/drug effects , Brain/drug effects , Brain/metabolism , Brain/pathology , Chromatography, Liquid , Dermatan Sulfate/cerebrospinal fluid , Disease Models, Animal , Heparitin Sulfate/genetics , Humans , Iduronate Sulfatase/genetics , Mice , Mice, Knockout , Mucopolysaccharidosis II/drug therapy , Mucopolysaccharidosis II/genetics , Mucopolysaccharidosis II/pathology , Nervous System Diseases/pathology , Receptors, Transferrin/genetics , Tandem Mass Spectrometry
2.
Gene ; 575(2 Pt 3): 577-83, 2016 Jan 10.
Article in English | MEDLINE | ID: mdl-26390815

ABSTRACT

Epac (exchange protein activated by cyclic-AMP) 2 is a direct target of 3'-5'-cyclic adenosine monophosphate (cAMP) and is involved in cAMP-mediated signal transduction through activation of the Ras-like small GTPase Rap. Crystallographic analyses revealed that activation of Epac2 by cAMP is accompanied by dynamic structural changes. Epac2 is expressed mainly in brain, neuroendocrine and endocrine tissues, and is involved in diverse cellular functions in the tissues. In this review, we summarize the structure and function of Epac2. We also discuss the physiological and pathophysiological roles of Epac2, and the possibility of Epac2 as a therapeutic target.


Subject(s)
Cyclic AMP/metabolism , Guanine Nucleotide Exchange Factors/chemistry , Guanine Nucleotide Exchange Factors/metabolism , Animals , Brain/metabolism , Endocrine System/metabolism , Humans , Myocardium/metabolism , Protein Conformation , Signal Transduction , ras Proteins/metabolism
3.
Diabetes ; 64(4): 1262-72, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25315008

ABSTRACT

Incretin-related drugs and sulfonylureas are currently used worldwide for the treatment of type 2 diabetes. We recently found that Epac2A, a cAMP binding protein having guanine nucleotide exchange activity toward Rap, is a target of both incretin and sulfonylurea. This suggests the possibility of interplay between incretin and sulfonylurea through Epac2A/Rap1 signaling in insulin secretion. In this study, we examined the combinatorial effects of incretin and various sulfonylureas on insulin secretion and activation of Epac2A/Rap1 signaling. A strong augmentation of insulin secretion by combination of GLP-1 and glibenclamide or glimepiride, which was found in Epac2A(+/+) mice, was markedly reduced in Epac2A(-/-) mice. In contrast, the combinatorial effect of GLP-1 and gliclazide was rather mild, and the effect was not altered by Epac2A ablation. Activation of Rap1 was enhanced by the combination of an Epac-selective cAMP analog with glibenclamide or glimepiride but not gliclazide. In diet-induced obese mice, ablation of Epac2A reduced the insulin secretory response to coadministration of the GLP-1 receptor agonist liraglutide and glimepiride. These findings clarify the critical role of Epac2A/Rap1 signaling in the augmenting effect of incretin and sulfonylurea on insulin secretion and provide the basis for the effects of combination therapies of incretin-related drugs and sulfonylureas.


Subject(s)
Glucagon-Like Peptide 1/pharmacology , Guanine Nucleotide Exchange Factors/metabolism , Insulin/metabolism , Signal Transduction/drug effects , Sulfonylurea Compounds/pharmacology , rap1 GTP-Binding Proteins/metabolism , Animals , Calcium/metabolism , Incretins/pharmacology , Insulin Secretion , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Mice , Signal Transduction/physiology
4.
Cell Rep ; 9(2): 661-73, 2014 Oct 23.
Article in English | MEDLINE | ID: mdl-25373904

ABSTRACT

Incretins, hormones released by the gut after meal ingestion, are essential for maintaining systemic glucose homeostasis by stimulating insulin secretion. The effect of incretins on insulin secretion occurs only at elevated glucose concentrations and is mediated by cAMP signaling, but the mechanism linking glucose metabolism and cAMP action in insulin secretion is unknown. We show here, using a metabolomics-based approach, that cytosolic glutamate derived from the malate-aspartate shuttle upon glucose stimulation underlies the stimulatory effect of incretins and that glutamate uptake into insulin granules mediated by cAMP/PKA signaling amplifies insulin release. Glutamate production is diminished in an incretin-unresponsive, insulin-secreting ß cell line and pancreatic islets of animal models of human diabetes and obesity. Conversely, a membrane-permeable glutamate precursor restores amplification of insulin secretion in these models. Thus, cytosolic glutamate represents the elusive link between glucose metabolism and cAMP action in incretin-induced insulin secretion.


Subject(s)
Cyclic AMP/metabolism , Exocytosis , Glucose/metabolism , Glutamic Acid/metabolism , Incretins/pharmacology , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Animals , Cell Line, Tumor , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/metabolism , Cytoplasm/metabolism , Incretins/metabolism , Insulin-Secreting Cells/drug effects , Male , Metabolome , Mice , Rats , Rats, Wistar , Secretory Vesicles/metabolism , Signal Transduction
5.
Sci Signal ; 6(298): ra94, 2013 Oct 22.
Article in English | MEDLINE | ID: mdl-24150255

ABSTRACT

Sulfonylureas are widely used drugs for treating insulin deficiency in patients with type 2 diabetes. Sulfonylureas bind to the regulatory subunit of the pancreatic ß cell potassium channel that controls insulin secretion. Sulfonylureas also bind to and activate Epac2A, a member of the Epac family of cyclic adenosine monophosphate (cAMP)-binding proteins that promote insulin secretion through activation of the Ras-like guanosine triphosphatase Rap1. Using molecular docking simulation, we identified amino acid residues in one of two cyclic nucleotide-binding domains, cNBD-A, in Epac2A predicted to mediate the interaction with sulfonylureas. We confirmed the importance of the identified residues by site-directed mutagenesis and analysis of the response of the mutants to sulfonylureas using two assays: changes in fluorescence resonance energy transfer (FRET) of an Epac2A-FRET biosensor and direct sulfonylurea-binding experiments. These residues were also required for the sulfonylurea-dependent Rap1 activation by Epac2A. Binding of sulfonylureas to Epac2A depended on the concentration of cAMP and the structures of the drugs. Sulfonylureas and cAMP cooperatively activated Epac2A through binding to cNBD-A and cNBD-B, respectively. Our data suggest that sulfonylureas stabilize Epac2A in its open, active state and provide insight for the development of drugs that target Epac2A.


Subject(s)
Cyclic AMP/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Hypoglycemic Agents/metabolism , Sulfonylurea Compounds/metabolism , Animals , Binding Sites/genetics , Binding, Competitive/genetics , COS Cells , Cell Line, Tumor , Cells, Cultured , Chlorocebus aethiops , Cyclic AMP/chemistry , Fluorescence Resonance Energy Transfer , Guanine Nucleotide Exchange Factors/chemistry , Guanine Nucleotide Exchange Factors/genetics , Histidine/chemistry , Histidine/genetics , Histidine/metabolism , Hypoglycemic Agents/chemistry , Mice , Mice, Knockout , Models, Molecular , Molecular Structure , Mutagenesis, Site-Directed , Mutation , Protein Stability , Protein Structure, Tertiary , Sulfonylurea Compounds/chemistry
6.
J Biol Chem ; 288(36): 25851-25864, 2013 Sep 06.
Article in English | MEDLINE | ID: mdl-23867458

ABSTRACT

Actin dynamics in pancreatic ß-cells is involved in insulin secretion. However, the molecular mechanisms of the regulation of actin dynamics by intracellular signals in pancreatic ß-cells and its role in phasic insulin secretion are largely unknown. In this study, we elucidate the regulation of actin dynamics by neuronal Wiskott-Aldrich syndrome protein (N-WASP) and cofilin in pancreatic ß-cells and demonstrate its role in glucose-induced insulin secretion (GIIS). N-WASP, which promotes actin polymerization through activation of the actin nucleation factor Arp2/3 complex, was found to be activated by glucose stimulation in insulin-secreting clonal pancreatic ß-cells (MIN6-K8 ß-cells). Introduction of a dominant-negative mutant of N-WASP, which lacks G-actin and Arp2/3 complex-binding region VCA, into MIN6-K8 ß-cells or knockdown of N-WASP suppressed GIIS, especially the second phase. We also found that cofilin, which severs F-actin in its dephosphorylated (active) form, is converted to the phosphorylated (inactive) form by glucose stimulation in MIN6-K8 ß-cells, thereby promoting F-actin remodeling. In addition, the dominant-negative mutant of cofilin, which inhibits activation of endogenous cofilin, or knockdown of cofilin reduced the second phase of GIIS. However, the first phase of GIIS occurs in the G-actin predominant state, in which cofilin activity predominates over N-WASP activity. Thus, actin dynamics regulated by the balance of N-WASP and cofilin activities determines the biphasic response of GIIS.


Subject(s)
Actin Depolymerizing Factors/metabolism , Glucose/pharmacology , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Sweetening Agents/pharmacology , Wiskott-Aldrich Syndrome Protein, Neuronal/metabolism , Actin Depolymerizing Factors/genetics , Actin-Related Protein 2-3 Complex/genetics , Actin-Related Protein 2-3 Complex/metabolism , Actins/genetics , Actins/metabolism , Animals , Cell Line , Gene Knockdown Techniques , Humans , Insulin/genetics , Insulin Secretion , Insulin-Secreting Cells/cytology , Male , Mice , Wiskott-Aldrich Syndrome Protein, Neuronal/genetics
7.
Nat Med ; 19(5): 567-75, 2013 May.
Article in English | MEDLINE | ID: mdl-23542788

ABSTRACT

Glucagon-like peptide-1 receptor (GLP-1R) agonists exert antihypertensive actions through incompletely understood mechanisms. Here we demonstrate that cardiac Glp1r expression is localized to cardiac atria and that GLP-1R activation promotes the secretion of atrial natriuretic peptide (ANP) and a reduction of blood pressure. Consistent with an indirect ANP-dependent mechanism for the antihypertensive effects of GLP-1R activation, the GLP-1R agonist liraglutide did not directly increase the amount of cyclic GMP (cGMP) or relax preconstricted aortic rings; however, conditioned medium from liraglutide-treated hearts relaxed aortic rings in an endothelium-independent, GLP-1R-dependent manner. Liraglutide did not induce ANP secretion, vasorelaxation or lower blood pressure in Glp1r(-/-) or Nppa(-/-) mice. Cardiomyocyte GLP-1R activation promoted the translocation of the Rap guanine nucleotide exchange factor Epac2 (also known as Rapgef4) to the membrane, whereas Epac2 deficiency eliminated GLP-1R-dependent stimulation of ANP secretion. Plasma ANP concentrations were increased after refeeding in wild-type but not Glp1r(-/-) mice, and liraglutide increased urine sodium excretion in wild-type but not Nppa(-/-) mice. These findings define a gut-heart GLP-1R-dependent and ANP-dependent axis that regulates blood pressure.


Subject(s)
Atrial Natriuretic Factor/metabolism , Blood Pressure , Gene Expression Regulation , Guanine Nucleotide Exchange Factors/metabolism , Receptors, Glucagon/metabolism , Animals , Aorta/metabolism , Aorta/pathology , Cyclic GMP/metabolism , Diabetes Mellitus, Type 2/drug therapy , Endothelium, Vascular/metabolism , Glucagon-Like Peptide 1/analogs & derivatives , Glucagon-Like Peptide 1/pharmacology , Glucagon-Like Peptide-1 Receptor , Liraglutide , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Natriuretic Peptide, C-Type/genetics , Perfusion , Protein Precursors/genetics , Vasodilation
9.
J Clin Invest ; 121(6): 2118-25, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21633180

ABSTRACT

Insulin secretion is a highly dynamic process regulated by various factors including nutrients, hormones, and neuronal inputs. The dynamics of insulin secretion can be studied at different levels: the single ß cell, pancreatic islet, whole pancreas, and the intact organism. Studies have begun to analyze cellular and molecular mechanisms underlying dynamics of insulin secretion. This review focuses on our current understanding of the dynamics of insulin secretion in vitro and in vivo and discusses their clinical relevance.


Subject(s)
Diabetes Mellitus/physiopathology , Insulin/metabolism , Islets of Langerhans/metabolism , Obesity/physiopathology , Animals , Circadian Rhythm/physiology , Cyclic AMP/physiology , Cytoplasmic Granules/metabolism , Diabetes Mellitus/drug therapy , Eating/physiology , Glucose Metabolism Disorders/physiopathology , Guanine Nucleotide Exchange Factors/agonists , Guanine Nucleotide Exchange Factors/physiology , Humans , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Incretins/physiology , Insulin Resistance/physiology , Insulin Secretion , Models, Biological , Obesity/drug therapy , Potassium Channels/drug effects , Potassium Channels/physiology , Secretory Rate , Signal Transduction/drug effects , Signal Transduction/physiology
10.
Nihon Rinsho ; 69(5): 803-7, 2011 May.
Article in Japanese | MEDLINE | ID: mdl-21595262

ABSTRACT

Incretin hormones GIP(glucose-dependent insulinotropic polypeptide) and GLP-1 (glucagon-like peptide-1) improve glycemic control by potentiating glucose-induced insulin secretion in pancreatic beta-cells and also have beneficial effects on appetite control and body weight. In response to food ingestion, GIP and GLP-1 are secreted from enteroendocrine K- and L-cells, respectively. In these cells, it is shown that a variety of molecular sensors are involved in the detection of carbohydrates, lipids, and proteins. In view of development of new incretin-related drugs, these sensors are attractive targets to enhance the endogenous pools of incretins.


Subject(s)
Enteroendocrine Cells/metabolism , Gastric Inhibitory Polypeptide/metabolism , Glucagon-Like Peptide 1/metabolism , Humans
11.
Cell Metab ; 12(2): 117-29, 2010 Aug 04.
Article in English | MEDLINE | ID: mdl-20674857

ABSTRACT

Insulin secretion is essential for maintenance of glucose homeostasis, but the mechanism of insulin granule exocytosis, the final step of insulin secretion, is largely unknown. Here, we investigated the role of Rim2alpha in insulin granule exocytosis, including the docking, priming, and fusion steps. We found that interaction of Rim2alpha and Rab3A is required for docking, which is considered a brake on fusion events, and that docking is necessary for K(+)-induced exocytosis, but not for glucose-induced exocytosis. Furthermore, we found that dissociation of the Rim2alpha/Munc13-1 complex by glucose stimulation activates Syntaxin1 by Munc13-1, indicating that Rim2alpha primes insulin granules for fusion. Thus, Rim2alpha determines docking and priming states in insulin granule exocytosis depending on its interacting partner, Rab3A or Munc13-1, respectively. Because Rim2alpha(-/-) mice exhibit impaired secretion of various hormones stored as dense-core granules, including glucose-dependent insulinotropic polypeptide, growth hormone, and epinephrine, Rim2alpha plays a critical role in exocytosis of these dense-core granules.


Subject(s)
Exocytosis/physiology , GTP-Binding Proteins/metabolism , Insulin/metabolism , rab3 GTP-Binding Proteins/metabolism , Animals , GTP-Binding Proteins/deficiency , GTP-Binding Proteins/genetics , Insulin Secretion , Insulin-Secreting Cells/metabolism , Mice , Nerve Tissue Proteins/metabolism , Potassium/metabolism , Secretory Vesicles/metabolism , Syntaxin 1/metabolism , rab3 GTP-Binding Proteins/deficiency , rab3 GTP-Binding Proteins/genetics , rab3A GTP-Binding Protein/metabolism
12.
Article in English | MEDLINE | ID: mdl-20551594

ABSTRACT

Pancreatic beta-cells play a central role in the maintenance glucose homeostasis by secreting insulin, a key hormone that regulates blood glucose levels. Dysfunction of the beta-cells and/or a decrease in the beta-cell mass are associated closely with the pathogenesis and pathophysiology of diabetes mellitus, a major metabolic disease that is rapidly increasing worldwide. Clarification of the mechanisms of insulin secretion and beta-cell fate provides a basis for the understanding of diabetes and its better treatment. In this review, we discuss cell signaling critical for the insulin secretory function based on our recent studies.


Subject(s)
Diabetes Mellitus/pathology , Diabetes Mellitus/therapy , Insulin-Secreting Cells/pathology , Signal Transduction , Animals , Cyclic AMP/metabolism , Diabetes Mellitus/metabolism , Humans , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Pancreas, Exocrine/pathology
13.
Cell Metab ; 11(6): 543-553, 2010 Jun 09.
Article in English | MEDLINE | ID: mdl-20519125

ABSTRACT

Glucagon secretion is inhibited by glucagon-like peptide-1 (GLP-1) and stimulated by adrenaline. These opposing effects on glucagon secretion are mimicked by low (1-10 nM) and high (10 muM) concentrations of forskolin, respectively. The expression of GLP-1 receptors in alpha cells is <0.2% of that in beta cells. The GLP-1-induced suppression of glucagon secretion is PKA dependent, is glucose independent, and does not involve paracrine effects mediated by insulin or somatostatin. GLP-1 is without much effect on alpha cell electrical activity but selectively inhibits N-type Ca(2+) channels and exocytosis. Adrenaline stimulates alpha cell electrical activity, increases [Ca(2+)](i), enhances L-type Ca(2+) channel activity, and accelerates exocytosis. The stimulatory effect is partially PKA independent and reduced in Epac2-deficient islets. We propose that GLP-1 inhibits glucagon secretion by PKA-dependent inhibition of the N-type Ca(2+) channels via a small increase in intracellular cAMP ([cAMP](i)). Adrenaline stimulates L-type Ca(2+) channel-dependent exocytosis by activation of the low-affinity cAMP sensor Epac2 via a large increase in [cAMP](i).


Subject(s)
Calcium Channels, L-Type/metabolism , Calcium Channels, N-Type/metabolism , Epinephrine/metabolism , Exocytosis , Glucagon-Like Peptide 1/metabolism , Glucagon/metabolism , Animals , Carrier Proteins/metabolism , Cells, Cultured , Colforsin/pharmacology , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Islets of Langerhans/cytology , Membrane Potentials , Mice , Mice, Inbred C57BL
14.
J Diabetes Investig ; 1(1-2): 37-9, 2010 Apr 22.
Article in English | MEDLINE | ID: mdl-24843406

ABSTRACT

Sulfonylureas (SU), commonly used in the treatment of type 2 diabetes mellitus (T2DM), stimulate insulin secretion by inhibiting adenosine triphosphate (ATP)-sensitive K(+) (KATP) channels in pancreatic ß-cells. SU are now known to also activate cyclic adenosine monophosphate (cAMP) sensor Epac2 (cAMP-GEFII) to Rap1 signaling, which promotes insulin secretion. The different effects of various SU on Epac2/Rap1 signaling, as well as KATP channels in different tissues, underlie the diverse pancreatic and extra-pancreatic actions of SU. (J Diabetes Invest, doi: 10.1111/j.2040-1124.2010.00014.x, 2010).

15.
J Diabetes Investig ; 1(4): 137-42, 2010 Aug 02.
Article in English | MEDLINE | ID: mdl-24843422

ABSTRACT

Incretin/cyclic adenosine monophosphate (cAMP) signaling is critical for potentiation of insulin secretion. Although several cell lines of pancreatic ß-cells are currently available, there are no cell lines suitable for investigation of incretin/cAMP signaling. In the present study, we have newly established pancreatic ß-cell lines (named MIN6-K) from the IT6 mouse, which develops insulinoma. MIN6-K8 cells respond to both glucose and incretins, such as glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP), as is the case in pancreatic islets, whereas MIN6-K20 cells respond to glucose, but not to incretins. Despite the difference in incretin-potentiated insulin secretion between these two cell lines, the accumulation of cAMP after stimulation of GLP-1 is comparable in these cells. Interestingly, we also found that incretin responsiveness is drastically induced by the formation of pseudoislets from MIN6-K20 cells to a level comparable to that of pancreatic islets. Thus, these cell lines are useful for studying incretin/cAMP signaling in ß-cells. (J Diabetes Invest, doi: 10.1111/j.2040-1124.2010.00026.x, 2010).

16.
Science ; 325(5940): 607-10, 2009 Jul 31.
Article in English | MEDLINE | ID: mdl-19644119

ABSTRACT

Epac2, a guanine nucleotide exchange factor for the small guanosine triphosphatase Rap1, is activated by adenosine 3',5'-monophosphate. Fluorescence resonance energy transfer and binding experiments revealed that sulfonylureas, widely used antidiabetic drugs, interact directly with Epac2. Sulfonylureas activated Rap1 specifically through Epac2. Sulfonylurea-stimulated insulin secretion was reduced both in vitro and in vivo in mice lacking Epac2, and the glucose-lowering effect of the sulfonylurea tolbutamide was decreased in these mice. Epac2 thus contributes to the effect of sulfonylureas to promote insulin secretion. Because Epac2 is also required for the action of incretins, gut hormones crucial for potentiating insulin secretion, it may be a promising target for antidiabetic drug development.


Subject(s)
Carrier Proteins/metabolism , Cyclic AMP/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Hypoglycemic Agents/metabolism , Sulfonylurea Compounds/metabolism , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Animals , Blood Glucose/analysis , COS Cells , Carrier Proteins/genetics , Cell Line , Chlorocebus aethiops , Fluorescence Resonance Energy Transfer , Glucose/administration & dosage , Glyburide/metabolism , Glyburide/pharmacology , Guanine Nucleotide Exchange Factors/genetics , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/pharmacology , Insulin/blood , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/metabolism , Mice , Mice, Inbred C57BL , Sulfonylurea Compounds/chemistry , Sulfonylurea Compounds/pharmacology , Tolbutamide/metabolism , Tolbutamide/pharmacology , rap1 GTP-Binding Proteins/metabolism
17.
Genes Cells ; 14(4): 445-56, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19335615

ABSTRACT

Rab GTPases and their effectors play important roles in membrane trafficking between cellular compartments in eukaryotic cells. In the present study, we examined the roles of Rab11B and its effectors in insulin secretion in pancreatic beta-cells. In the mouse insulin-secreting cell line MIN6, Rab11 was co-localized with insulin-containing granules, and over-expression of the GTP- or the GDP-bound form of Rab11B significantly inhibited regulated secretion, indicating involvement of Rab11B in regulated insulin secretion. To determine the downstream signal of Rab11-mediated insulin secretion, we examined the effects of various Rab11-interacting proteins on insulin secretion, and found that Rip11 is involved in cAMP-potentiated insulin secretion but not in glucose-induced insulin secretion. Analyses by immunocytochemistry and subcellular fractionation revealed Rip11 to be co-localized with insulin granules. The inhibitory effect of the Rip11 mutant was not altered in MIN6 cells lacking Epac2, which mediates protein kinase A (PKA)-independent potentiation of insulin secretion, compared with wild-type MIN6 cells. In addition, Rip11 was found to be phosphorylated by PKA in MIN6 cells. The present study shows that both Rab11 and its effector Rip11 participate in insulin granule exocytosis and that Rip11, as a substrate of PKA, regulates the potentiation of exocytosis by cAMP in pancreatic beta-cells.


Subject(s)
Carrier Proteins/metabolism , Exocytosis , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Mitochondrial Proteins/metabolism , rab GTP-Binding Proteins/metabolism , 1-Methyl-3-isobutylxanthine/pharmacology , Animals , Carrier Proteins/genetics , Cell Line, Tumor , Colforsin/pharmacology , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/metabolism , Glucose/pharmacology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Immunoblotting , Insulin Secretion , Insulin-Secreting Cells/cytology , Insulinoma/metabolism , Insulinoma/pathology , Insulinoma/ultrastructure , Isoquinolines/pharmacology , Mice , Microscopy, Fluorescence , Microscopy, Immunoelectron , Mitochondrial Proteins/genetics , Okadaic Acid/pharmacology , Phosphorylation/drug effects , Protein Transport/drug effects , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sulfonamides/pharmacology , Transfection , rab GTP-Binding Proteins/genetics
18.
J Cell Physiol ; 219(3): 652-8, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19170062

ABSTRACT

cAMP is a well-known regulator of exocytosis, and cAMP-GEFII (Epac2) is involved in the potentiation of cAMP-dependent, PKA-independent regulated exocytosis in secretory cells. However, the mechanisms of its action are not fully understood. In the course of our study of Epac2 knockout mice, we identified a novel splicing variant of Epac2, which we designate Epac2B, while renaming the previously identified Epac2 Epac2A. Epac2B, which lacks the first cAMP-binding domain A in the N-terminus but has the second cAMP-binding domain B of Epac2A, possesses GEF activity towards Rap1, as was found for Epac2A. Immunocytochemical analysis revealed that exogenously introduced Epac2A into insulin-secreting MIN6 cells was localized near the plasma membrane, while Epac2B was found primarily in the cytoplasm. Interestingly, cAMP-binding domain A alone introduced into MIN6 cells was also localized near the plasma membrane. In MIN6 cells, Epac2A was involved in triggering hormone secretion by stimulation with 5.6 mM glucose plus 1 mM 8-Bromo-cAMP, but Epac2B was not. The addition of a membrane-targeting signal to the N-terminus of Epac2B was able to mimic the effect of Epac2A on hormone secretion. Thus, the present study indicates that the N-terminal cAMP-binding domain A of Epac2A plays a critical role in determining its subcellular localization and potentiating insulin secretion by cAMP. J. Cell. Physiol. 219: 652-658, 2009. (c) 2009 Wiley-Liss, Inc.


Subject(s)
Carrier Proteins/chemistry , Carrier Proteins/metabolism , Cyclic AMP/metabolism , Guanine Nucleotide Exchange Factors/chemistry , Guanine Nucleotide Exchange Factors/metabolism , Adrenal Glands/metabolism , Alternative Splicing , Amino Acid Sequence , Animals , Binding Sites , Carrier Proteins/genetics , Cell Line , Cerebral Cortex/metabolism , Gene Expression , Guanine Nucleotide Exchange Factors/genetics , Mice , Mice, Knockout , Molecular Sequence Data , Protein Structure, Tertiary , Signal Transduction , Subcellular Fractions/metabolism , rap1 GTP-Binding Proteins/metabolism
19.
Biomed Res ; 29(2): 85-91, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18480549

ABSTRACT

The exocyst is an octameric complex involved in docking or tethering of secretory vesicles to fusion sites of the plasma membrane. Sec6 is the core subunit of the exocyst complex. Here we identify an isoform of Sec6, deposited as Exocyst complex component 3-like (Exoc3l) in the database, by in silico screening using rat Sec6 as a probe. The amino acid sequence of Exoc3l has 31% identity and 53% similarity with that of Sec6. RT-PCR analysis reveals that Exoc3l is expressed in insulin-secreting MIN6 cells as well as in various tissues including pancreatic islets and brain. In co-immunoprecipitation experiments, Exoc3l was found to interact with Sec5, Sec8, and Sec10, all of which are binding partners of Sec6 in the exocyst complex. Furthermore, overexpression of a deletion mutant of Exoc3l in MIN6 cells suppressed glucose-stimulated secretion. These results suggest that Exoc3l is involved in regulated exocytosis of insulin granules.


Subject(s)
Carrier Proteins/chemistry , Carrier Proteins/physiology , Insulin/metabolism , Structural Homology, Protein , Vesicular Transport Proteins/chemistry , Vesicular Transport Proteins/physiology , Amino Acid Sequence , Animals , Brain/metabolism , Exocytosis/physiology , Humans , Insulin Secretion , Islets of Langerhans/metabolism , Membrane Proteins , Mice , Molecular Sequence Data , Organ Specificity/physiology , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Protein Isoforms/physiology , Vesicular Transport Proteins/metabolism
20.
Proc Natl Acad Sci U S A ; 104(49): 19333-8, 2007 Dec 04.
Article in English | MEDLINE | ID: mdl-18040047

ABSTRACT

cAMP is well known to regulate exocytosis in various secretory cells, but the precise mechanism of its action remains unknown. Here, we examine the role of cAMP signaling in the exocytotic process of insulin granules in pancreatic beta cells. Although activation of cAMP signaling alone does not cause fusion of the granules to the plasma membrane, it clearly potentiates both the first phase (a prompt, marked, and transient increase) and the second phase (a moderate and sustained increase) of glucose-induced fusion events. Interestingly, all granules responsible for this potentiation are newly recruited and immediately fused to the plasma membrane without docking (restless newcomer). Importantly, cAMP-potentiated fusion events in the first phase of glucose-induced exocytosis are markedly reduced in mice lacking the cAMP-binding protein Epac2 (Epac2(ko/ko)). In addition, the small GTPase Rap1, which is activated by cAMP specifically through Epac2 in pancreatic beta cells, mediates cAMP-induced insulin secretion in a protein kinase A-independent manner. We also have developed a simulation model of insulin granule movement in which potentiation of the first phase is associated with an increase in the insulin granule density near the plasma membrane. Taken together, these data indicate that Epac2/Rap1 signaling is essential in regulation of insulin granule dynamics by cAMP, most likely by controlling granule density near the plasma membrane.


Subject(s)
Carrier Proteins/physiology , Cyclic AMP/physiology , Cytoplasmic Granules/metabolism , Exocytosis , Guanine Nucleotide Exchange Factors/physiology , Insulin/metabolism , rap1 GTP-Binding Proteins/metabolism , Animals , Carrier Proteins/genetics , Cell Line , Cytoplasmic Granules/chemistry , Cytoplasmic Granules/drug effects , Exocytosis/drug effects , Glucose/pharmacology , Guanine Nucleotide Exchange Factors/genetics , Humans , Insulin/analysis , Insulin Secretion , Mice , Mice, Knockout , Potassium/pharmacology , Signal Transduction , rap1 GTP-Binding Proteins/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...