PROPER GLUCOSE HOMEOSTASIS is critically dependent

Similar documents
NIH Public Access Author Manuscript Diabetologia. Author manuscript; available in PMC 2014 February 01.

Perspectives Home > Literature > Perspectives > Holz et al., pp. pe2

The glucoincretin hormone glucagon-like peptide-1

A novel role for vitamin D: modulation of expression and function of the local renin angiotensin system in mouse pancreatic islets

7/31/2009. G.Y. Prince Used Cars 10 am Los Angelos, CA Mullholland Drive..later that day. Would you buy a car without taking it for a spin first?

Diabetes: What is the scope of the problem?

The Effects of Exenatide on the Autoimmune Development of Diabetes. A Senior Honors Thesis

Heart disease is frequently associated with elevated wall

Glucagon-like peptide 1 (GLP-1) is secreted from

Phospho-AKT Sampler Kit

General Laboratory methods Plasma analysis: Gene Expression Analysis: Immunoblot analysis: Immunohistochemistry:

islets scored 1 week month months

Receptor mediated Signal Transduction

AT1 RECEPTOR BLOCKADE ATTENUATES INSULIN RESISTANCE AND MYOCARDIAL REMODELING IN RATS WITH DIET-INDUCED OBESITY

Glucagon-like peptide-1 treatment delays the onset of diabetes in 8 week-old db/db mice

Supporting Information

Inflammation & Type 2 Diabetes Prof. Marc Y. Donath

Cell Signaling part 2

Discussion & Conclusion

Abbreviations: P- paraffin-embedded section; C, cryosection; Bio-SA, biotin-streptavidin-conjugated fluorescein amplification.

adenosine triphosphate-sensitive K[ Citation Journal of Diabetes Investigation ( modifications or adaptations are ma

Week 3, Lecture 5a. Pathophysiology of Diabetes. Simin Liu, MD, ScD

Defects in glucose utilization & GLP-1

Pancreatic Insulinoma Presenting. with Episodes of Hypoinsulinemic. Hypoglycemia in Elderly ---- A Case Report

Supplementary Figure 1. DJ-1 modulates ROS concentration in mouse skeletal muscle.

Autonomic regulation of islet hormone secretion

Growth and Differentiation Phosphorylation Sampler Kit

Introduction. up regulated include genes associated with stress response, DNA repair and

KEY CONCEPT QUESTIONS IN SIGNAL TRANSDUCTION

Reduced expression of the murine p85α subunit of phosphoinositide 3-kinase improves insulin signaling and ameliorates diabetes

Insights into insulin resistance and type 2 diabetes from knockout mouse models

Supplementary Materials for

Div. of Endocrinology and Metabolism Konyang University, School of Medicine. Byung-Joon Kim M.D., Ph.D.

Goals and Challenges of Communication. Communication and Signal Transduction. How Do Cells Communicate?

G-Protein Signaling. Introduction to intracellular signaling. Dr. SARRAY Sameh, Ph.D

Figure S1. Body composition, energy homeostasis and substrate utilization in LRH-1 hep+/+ (white bars) and LRH-1 hep-/- (black bars) mice.

Principles of Genetics and Molecular Biology

Glucose Homeostasis. Liver. Glucose. Muscle, Fat. Pancreatic Islet. Glucose utilization. Glucose production, storage Insulin Glucagon

Insulin Resistance. Biol 405 Molecular Medicine

Neuroprotective properties of GLP-1 - a brief overview. Michael Gejl Jensen, MD Dept. Of Pharmacology, AU

Making Mature Human Islet Cells from Stem Cells to Model Disease and Treat Diabetes

Journal Club WS 2012/13 Stefanie Nickl

Original Article Protective effect of ginsenoside Rg1 on isoproterenol-induced acute myocardial ischemia in rats

Prior in vitro exposure to GLP-1 with or without GIP can influence the subsequent beta cell responsiveness

SUPPLEMENTARY INFORMATION

Title Modified Western blotting for insulin and other diabetes-associated peptide hormones

Management of Type 2 Diabetes

A dual PI3 kinase/mtor inhibitor reveals emergent efficacy in glioma

UNIVERSITY OF PNG SCHOOL OF MEDICINE AND HEALTH SCIENCES DIVISION OF BASIC MEDICAL SCIENCES DISCIPLINE OF BIOCHEMISTRY AND MOLECULAR BIOLOGY

Endocannabinoid-activated Nlrp3 inflammasome in infiltrating macrophages mediates β- cell loss in type 2 diabetes

Article. Reference. Effect of insulin on glucose- and arginine-stimulated somatostatin secretion from the isolated perfused rat pancreas

Cellular Signaling Pathways. Signaling Overview

BMP6 treatment compensates for the molecular defect and ameliorates hemochromatosis in Hfe knockout mice

Effect of macronutrients and mixed meals on incretin hormone secretion and islet cell function

Therapeutic strategy to reduce Glucagon secretion

AMPK Phosphorylation Assay Kit

AP VP DLP H&E. p-akt DLP

Signal Transduction Pathways. Part 2

Disruption of Insulin Receptor Substrate 2 Causes Type 2 Diabetes Because of Liver Insulin Resistance and Lack of Compensatory

28 Regulation of Fasting and Post-

Effect of Glucagon-Like Peptide-1 on - and -Cell Function in C-Peptide-Negative Type 1 Diabetic Patients

Despite substantial advances in our understanding

ANUMBER OF gut peptide hormones exhibit diverse

Hormones. Prof. Dr. Volker Haucke Institut für Chemie-Biochemie Takustrasse 6

Supplementary Table 1. The primers used for quantitative RT-PCR. Gene name Forward (5 > 3 ) Reverse (5 > 3 )

Final Review Sessions. 3/16 (FRI) 126 Wellman (4-6 6 pm) 3/19 (MON) 1309 Surge 3 (4-6 6 pm) Office Hours

Continuous stimulation of human glucagon-like peptide-1 (7 36) amide in a mouse model (NOD) delays onset of autoimmune type 1 diabetes

Supplementary Figure 1

Exendin-4-mediated cell proliferation in beta-cells: focusing on the transcription factors

Impact of transcription factor 7-like 2 (TCF7L2) on pancreatic islet function and morphology in mice and men.

Crosstalk between Adiponectin and IGF-IR in breast cancer. Prof. Young Jin Suh Department of Surgery The Catholic University of Korea

MPB333:Molecular Endocrinology of Obesity and Diabetes

Maternal diet induced micrornas and mtor underlie b cell dysfunction in offspring

Scope. History. History. Incretins. Incretin-based Therapy and DPP-4 Inhibitors

Part-4. Cell cycle regulatory protein 5 (Cdk5) A novel target of ERK in Carb induced cell death

What systems are involved in homeostatic regulation (give an example)?

Immunostaining was performed on tumor biopsy samples arranged in a tissue-microarray format or on

The Mediterranean Diet: HOW and WHY It Works So Well for T2DM

THE OBSERVATION, that oral glucose administration

SUPPLEMENTARY INFORMATION

Supplementary Table 1. Primer sequences for conventional RT-PCR on mouse islets

GENERAL CHARACTERISTICS OF THE ENDOCRINE SYSTEM FIGURE 17.1

Role of endogenous ROS in pancreatic β-cell dysfunction

Chapter 20. Cell - Cell Signaling: Hormones and Receptors. Three general types of extracellular signaling. endocrine signaling. paracrine signaling

Potentiation of Glucose-stimulated Insulin Secretion by the GPR40 PLC TRPC

Megan Lawless. Journal Club. January 20 th, 2011

Signal transduction of insulin

The PI3K/AKT axis. Dr. Lucio Crinò Medical Oncology Division Azienda Ospedaliera-Perugia. Introduction

Insulin resistance and pancreatic b cell failure

SUPPLEMENTARY INFORMATION

NIH Public Access Author Manuscript Diabetes Res Clin Pract. Author manuscript; available in PMC 2011 March 1.

Reduced Insulin Signaling and Endoplasmic Reticulum Stress Act Synergistically to Deteriorate Pancreatic β Cell Function

Signal Transduction Cascades

Type 2 DM in Adolescents: Use of GLP-1 RA. Objectives. Scope of Problem: Obesity. Background. Pathophysiology of T2DM

EAT TO LIVE: THE ROLE OF THE PANCREAS. Felicia V. Nowak, M.D., Ph.D. Ohio University COM 22 January, 2008

ARTICLE. C. Alarcon & B. Wicksteed & C. J. Rhodes

Glucagon-like peptide-1 (GLP-1), a 30 amino

Protection against doxorubicin-induced myocardial dysfunction in mice by cardiac-specific expression of carboxyl terminus of hsp70-interacting protein

Chapter 16: Endocrine System 1

Transcription:

0013-7227/04/$15.00/0 Endocrinology 145(9):4078 4083 Printed in U.S.A. Copyright 2004 by The Endocrine Society doi: 10.1210/en.2004-0028 Impaired Glucose-Stimulated Insulin Secretion, Enhanced Intraperitoneal Insulin Tolerance, and Increased -Cell Mass in Mice Lacking the p110 Isoform of Phosphoinositide 3-Kinase P. E. MACDONALD, J. W. JOSEPH, D. YAU, J. DIAO, Z. ASGHAR, F. DAI, G. Y. OUDIT, M. M. PATEL, P. H. BACKX, AND M. B. WHEELER University of Toronto (P.E.M., J.W.J., D.Y., J.D., Z.A., F.D., M.B.W.), Departments of Physiology and Medicine, Toronto, Canada; Lund University (P.E.M.), Department of Molecular and Cellular Physiology, Lund 221 84, Sweden; and Division of Cardiology (G.Y.O., M.M.P., P.H.B.), University Health Network and the Heart and Stroke Richard Lewar Centre of Excellence, Toronto, Canada M5S 3E2 Phosphoinositide 3-kinase (PI3 kinase) has been implicated in G protein-coupled receptor regulation of pancreatic -cell growth and glucose-stimulated insulin secretion. The G protein-activated p110 isoform of PI3 kinase was detected in insulinoma cells, mouse islets, and human islets. In 7- to 10- wk-old mice, knockout of p110 reduced the plasma insulin response to ip glucose injection and impaired first and second phase glucose-stimulated insulin secretion from pancreata perfused ex vivo. The p110 / mice responded to preinjection with the glucagon-like peptide-1 receptor agonist exendin 4, such that plasma glucose and insulin responses to ip glucose injection were not different from wild types. Mice PROPER GLUCOSE HOMEOSTASIS is critically dependent on an adequate pancreatic -cell mass that responds appropriately to increased blood glucose. Accordingly, defects in glucose-stimulated insulin secretion and an eventual decrease in -cell mass are important factors in the development and progression of type 2 diabetes (1). G protein-coupled receptors (GPCRs) such as those activated by glucagon-like peptide-1 (GLP-1) regulate -cell mass by promoting proliferation (2, 3) and preventing apoptosis (4, 5). Phosphoinositide 3-kinase (PI3 kinase) signaling is an important pathway mediating these effects (2, 6). In addition to its role in islet growth, numerous studies implicate PI3 kinase as a negative regulator of insulin secretion because its pharmacological inhibition (7, 8) and genetic reduction in mice (9) enhances secretion. PI3 kinase is comprised of both catalytic and regulatory subunits and has been extensively reviewed (10). The catalytic subunit of type 1A PI3 kinases, which are regulated by tyrosine kinase receptors, is one of three p110 isoforms (,, and ). These associate with one of five regulatory subunits; p50, p55, and p85, resulting from alternative splicing of a single gene; and p55 and p85. The only type 1B isoform Abbreviations: AUC, Area under the curve; GPCRs, G protein-coupled receptors; GLP-1, glucagon-like peptide-1; HI, human islets; p-akt, phosphorylated Akt; PI3 kinase, phosphoinositide 3-kinase. Endocrinology is published monthly by The Endocrine Society (http:// www.endo-society.org), the foremost professional society serving the endocrine community. lacking p110 were not diabetic and were only slightly glucose intolerant (ip glucose injection) compared with wild types, in part due to enhanced responsiveness to insulin as determined by an ip insulin tolerance test. Despite severely reduced insulin secretion in these animals, the p110 / mice had greater pancreatic insulin content, and an increased -cell mass due to -cell hypertrophy. These surprising results suggest that the G protein-coupled p110 isoform of PI3 kinase is not central to the development or maintenance of sufficient -cell mass but positively regulates glucose-stimulated insulin secretion. (Endocrinology 145: 4078 4083, 2004) of PI3 kinase, called p110, associates with the p101 regulatory subunit and is activated directly by GPCRs through an interaction with G. Like other PI3 kinases, p110 phosphorylates phosphatidylinositol 4,5-bisphosphate [PtdIns(4,5)P] to generate PtdIns(3,4,5)P in vivo and subsequently activates numerous downstream targets including 3-phosphoinositide-dependent kinase-1, Akt, protein kinase C, and p70s6 kinase. Activity of p110 has been demonstrated in insulinoma cells (11), whereas protein expression has been shown in human, dog, rat, and mouse pancreas by immunohistochemistry (12). Although the latter report suggests no expression of the protein in pancreatic islets, we show here that islet cells do indeed express p110. In contrast to the known effects of nonselective PI3 kinase inhibition and genetic reduction of type 1A PI3 kinase activity, we find that glucose-stimulated insulin secretion is blunted in mice lacking p110. The p110 mice are not diabetic, however, demonstrating enhanced responsiveness to ip insulin and increased -cell mass. This latter finding is surprising because G protein-activated PI3 kinase activity is thought to be a positive regulator of -cell proliferation (13). Therefore, the type 1B isoform p110 has an important role in islet function that is distinct from that of the type 1A PI3 kinases. Materials and Methods Animals and in vivo experiments Animal experiments were performed in accordance with University of Toronto guidelines. The generation of p110 / mice has been 4078

MacDonald et al. Brief Communications Endocrinology, September 2004, 145(9):4078 4083 4079 described (14). At 7 10 wk, male wild-type C57/Bl6 and p110 / mice did not differ in body weight (22.8 0.6 g and 24.5 0.4 g), and the p110 / mice were not diabetic. Intraperitoneal glucose (1.5 g/kg body weight) and insulin (0.75 U/kg body weight) tolerance tests were performed as described (15) on mice fasted overnight (15 18 h) and for 2 4 h, respectively. In some ip glucose tolerance experiments, exendin 4(5 g in PBS) was injected im (gluteal) 5 min before ip glucose. Blood glucose was measured with a Lifescan Elite glucose meter (Lifescan, Toronto, Ontario, Canada), and plasma insulin was assayed using a mouse insulin ELISA kit (Crystalchem, Chicago, IL). Glucagon was assayed after ip insulin injection (1 U/kg body weight) using a mouse glucagon RIA kit (Linco Research Inc., St. Charles, MO). Catecholamines were extracted from plasma collected after a 12-h fast using alumina, eluted, and assayed using HPLC (Waters Associates, Inc., Milford, MA) coupled with electrochemical detection (ESA Inc., Bedford, MA). Perfused pancreas Mice were fasted overnight (15 18 h), and the surgical procedure was similar to that described (16). The perfusate was a modified Krebs- Ringer-2% BSA-glucose-3% dextran solution gassed with 95% O 2 /5% CO 2 to achieve a ph of 7.4. Samples were assayed for insulin by RIA with a polyclonal insulin antibody from guinea pig (17) as described previously (15). -Cell mass and pancreatic insulin content Immunostaining for insulin and glucagon were performed as described (15). Images were collected at 4 or 40 magnification and analyzed using Image Pro software (MediaCybernetics, Carlsbad, CA). Insulin stained area, as a percentage of pancreas area, was multiplied by pancreas weight to calculate -cell mass. Islet area was calculated by dividing the insulin-positive area by the islet number for each tissue section. -Cell cross-sectional area was calculated by dividing the insulin-positive area by the number of -cell nuclei (3). -Cell density was determined by dividing the number of -cell nuclei by total pancreas area. Pancreatic insulin content was determined as described (15). Pancreata were collected after pancreas perfusion and stored at 80 C. Previous experiments confirmed no difference between this protocol and insulin content measured from pancreata taken without perfusion (15). RT-PCR and Western blot RT-PCR was performed on total RNA with 30 PCR cycles of 94 C for 30 sec, 56 C for 30 sec, and 72 C for 30 sec followed by a 10-min extension at 72 C. Primers specific to the p110 transcript, based on human sequences, were: forward 5 -ACTGCCTCAAGAACGGAGAA-3 and reverse 5 -CAGGAGGAAACGGTGGTCTA-3. Western blotting was performed as described (14) on whole cell lysates. MIN6 cells (passages 35 40) from S. Seino (Chiba University, Chuo-ku, Japan) were cultured as described previously (18). Human islets were obtained from the Juvenile Diabetes Research Foundation Human Islet Distribution Program courtesy J. Lakey (University of Alberta, Edmonton, Canada). Antibodies for p110 were from Santa Cruz Biotechnology Inc. (Santa Cruz, CA) and for glyceraldehyde-3-phosphate dehydrogenase were from Research Diagnostics Inc. (Flanders, NJ). Blotting for Akt and phosphorylated (p)-akt (Thr308) was performed on heart tissue from wild-type and p110 / mice 0, 10, and 30 min after ip insulin injection (1 U/kg body weight) and band densities were determined using Scion Image software (Scion Corp., Frederick, MD). Antibodies for Akt and p-akt (Thr308) were from Cell Signaling Technology (Beverly, MA). Statistics Curves were compared by one-way ANOVA and Bonferroni post test. Other data were compared using the unpaired Student s t test. P 0.05 was considered significant. Results Regulation of glucose homeostasis by p110 To examine a potential regulatory role for p110 in pancreatic islets, expression of this type 1B PI3 kinase was investigated in islets and insulin-secreting cells. Expression of p110 transcripts in total RNA from MIN-6 insulinoma cells, mouse islets (MI), and human islets (HI) was detected by RT-PCR using p110 -specific primers (Fig. 1A). No PCR product was obtained in water blank controls. Whereas heart [H( / )] and pancreas [P( / )] lysates from wild-type animals were positive for p110 protein expression by Western blot, expression could not be detected in pancreas from p110 / mice [P( / )] (Fig. 1A). Also, protein expression of p110 was detected in MIN-6 and HI lysates (Fig. 1A). Because both mouse and human islets express p110, a role for this PI3 kinase isoform in glucose homeostasis was investigated in p110 / mice. Fasting blood glucose levels were not different between wild-type (5.75 0.33 mm, n 11) and p110 / (6.03 0.27 mm, n 22) mice. The blood glucose excursion after an ip glucose load was greater in p110 / mice than in wild-type mice [area under the curve (AUC) of 1214.9 51.2 vs. 1029.0 60.1 mm min, n 22 and 11, P 0.05] (Fig. 1B). Before the glucose injection, plasma insulin levels were not different between wild-type (0.21 0.02 pg/ml, n 10) and p110 / mice (0.24 0.04 pg/ml, n 10). After the ip glucose load, plasma insulin levels in the p110 / mice were significantly impaired FIG. 1.A,Top, p110 transcripts were detected in total RNA from MIN-6 insulinoma cells, mouse islets (MI), and HI. No transcript was detectable in water blank controls (H 2 O). A, Bottom, p110 protein was detected by Western blot (two membranes) in mouse heart [H( / )], mouse pancreas [P( / )], MIN-6 cells and HI. Expression of p110 was not detected in pancreas from p110 / mice [P( / )]. B, Male mice (7 10 wk) were fasted overnight and injected ip with glucose. The blood glucose excursion was measured in wild-type (black) and p110 / (open) mice. The AUC (shown to the right) was greater in the p110 / mice. C, The plasma insulin response to the ip glucose injection was blunted in the p110 mice. The AUC values are shown at the right. *,P 0.05; **, P 0.01 compared with wild type.

4080 Endocrinology, September 2004, 145(9):4078 4083 MacDonald et al. Brief Communications (Fig. 1C) and the AUC of the insulin response in the / mice (69.3 9.9 pg ml 1 min, n 9) was decreased compared with wild types (145.8 19.8 pg ml 1 min, n 10, P 0.01) (Fig. 1C). Before ip insulin tolerance tests, blood glucose levels were not different between wild-type (8.01 0.28 mm,n 13) and p110 / mice (7.99 0.30 mm,n 15). After ip injection of insulin, blood glucose was decreased to a greater degree in the / mice compared with wild types ( AUC of 357.0 35.2 vs. 235.0 28.4 mm min, n 15 and 13, P 0.05) (Fig. 2A). Insulin-stimulated Akt phosphorylation (Thr308) in heart, at 10 or 30 min after insulin injection, was not significantly different between the wild-type and p110 / mice (Fig. 2B). In wild-type mice, plasma glucagon was elevated from 112.7 20.6 to 292.4 72.8 pg/ml (n 6, P 0.05) at 45 min after ip insulin (Fig. 2C). In the p110 / mice, plasma glucagon was depressed from 128.1 15.1 to 75.2 11.2 pg/ml (n 6, P 0.05) at 10 min but was exaggerated (799.1 162.9 pg/ml, n 6, P 0.001) compared with wild types (P 0.05) at 45 min (Fig. 2C). Fasting plasma epinephrine (3.84 0.57 and 4.28 0.87 nm, n 5) and norepinephrine (54.52 6.92 and 52.10 9.47 nm, n 5) levels were not different between the p110 / and wild-type mice (Fig. 2, D and E). Responsiveness of p110 / mice to exendin 4 Because the GLP-1 receptor can couple to PI3 kinase (2), the responsiveness of p110 / mice to the degradationresistant GLP-1 receptor agonist exendin 4 was examined. The blood glucose excursion during an ip glucose tolerance test was not different between wild-type and p110 / mice (not shown) after exendin 4 preinjection (5 g in PBS). The AUC values were 514.1 50.4 mm min (n 6) and 477.8 28.5 mm min (n 11), respectively, representing a significantly (P 0.001) enhanced glucose tolerance compared with untreated mice. Accordingly, after exendin 4 preinjection the plasma insulin response to the ip glucose tolerance test was not different between wild-type (AUC, 70.4 11.2 pg ml 1 min, n 7) and p110 / (AUC, 66.7 6.3 pg ml 1 min, n 7) mice. Pancreatic insulin content and -cell mass in p110 / mice PI3 kinase has been implicated in -cell proliferation, particularly in response to GLP-1 (13). Therefore, we examined whether the reduced plasma insulin response in p110 / mice was due to reduced -cell mass. Surprisingly, -cell mass in the / mice (1.65 0.21 mg, n 6) was greater than in wild types (0.86 0.23 mg, n 5, P 0.05) (Fig. 3B). Whereas the average islet number per pancreas area was not different (1.47 0.18 islets/mm 2,n 6 and 1.64 0.19 islets/mm 2,n 5) (Fig. 3C), average islet area was increased in p110 / (3088.6 320.8 m 2,n 6) vs. wild-type mice (1836.1 351.4 m 2,n 5, P 0.05) (Fig. 3D). Accordingly, the pancreatic insulin content of p110 / mice (91.5 12.2 ng/mg protein, n 7) was significantly greater than in wild types (54.7 5.5 ng/mg protein, n 7, P 0.05) (Fig. 3E). This resulted largely from -cell hypertrophy as demonstrated by an increase in -cell cross-sectional area from 150.0 5.4 m 2 (n 5) in the wild-type mice to 196.8 12.1 m 2 (n 5, P 0.01) in the p110 / mice (Fig. 3F), and no significant change in -cell density (22.4 4.8 and 25.0 3.7 -cells/mm 2 pancreas, n 5) (Fig. 3G). FIG. 2. A, Mice were fasted for 2 4 h and injected ip with insulin. Blood glucose (shown as the change in blood glucose levels) was reduced to a greater extent in the p110 / (open) mice compared with wild types (black). The corresponding change in AUC ( AUC) is shown at the right. B, Akt phorhporylation in heart was not significantly different between wild-type ( / ) and p110 / ( / ) mice 10 or 30 min after insulin injection. Blots are shown at the left and the fold increase in p-akt signal, compared with time 0 and normalized to total Akt, is shown at the right. Counterregulatory hormones were investigated in panel C. The plasma glucagon response after injection of insulin was depressed in the p110 / mice at 10 min but was exaggerated at 45 min. Fasting plasma norepinephrine and epinephrine levels were not different in wild-type vs. p110 / mice. *, P 0.05; **, P 0.01; and ***, P 0.001 compared wild type (A) or time 0. Ex vivo insulin secretion from p110 / pancreata Because -cell mass and pancreatic insulin content were not decreased in the p110 / mice, we examined insulin secretion using an ex vivo perfused pancreas model. Basal insulin secretion in the presence of substimulatory glucose (1.4 mm) was not different between groups, but secretion in response to glucose (13.4 mm) was decreased in the p110 / mice (Fig. 3A). Notably, first-phase insulin secretion was severely blunted (AUC from 77.0 11.0 to 36.3 8.6 pg ml 1 min, n 6, P 0.05), and second-phase secretion was also decreased (AUC from 107.0 13.7 to 66.0 11.2 pg ml 1 min, n 6, P 0.05) (Fig. 3, B and C). Insulin secretion in response to 20 mm arginine was not different

MacDonald et al. Brief Communications Endocrinology, September 2004, 145(9):4078 4083 4081 FIG. 4. Pancreata of wild-type and p110 / mice were perfused ex vivo. A, Wild-type mice (black) exhibited the classic bi-phasic secretion response to a step change in glucose (1.4 13.4 mm), whereas p110 / mice (open) showed a blunted response. The calculated AUC values for first phase (5 10 min) and second phase (10 22 min) secretion for wild-type and p110 / are shown in B and C. D, Wild-type and p110 / pancreata both responded to stimulation with 20 mm arginine. The AUC for the arginine response are shown in panel E. *,P 0.05; ***, P 0.001 compared with wild type. FIG. 3. Sections of wild-type and p110 / pancreata were stained for insulin and glucagon. A, Representative images obtained at 10 magnification. B, -Cell mass was increased in p110 / mice (open) compared with wild types (black). Although islet number (per pancreas area) was not different in wild-type vs. p110 / mice (C), the average islet area was increased in p110 / mice (D). Accordingly, total pancreatic insulin content (per microgram of total protein) was also increased in the p110 / mice (E). F and G, -Cell crosssectional area was increased in the p110 / mice, whereas -cell density (per pancreas area) was unchanged compared with wild types. *, P 0.05; **, P 0.01 compared with wild-type. between the / and wild-type preparations (AUC of 116.1 16.6 and 116.4 24.6 pg ml 1 min, respectively, n 6) (Fig. 4, D and E). Discussion The p110 isoform of PI3 kinase is activated by GPCRs thorough a direct interaction with the G -subunit (10). The initial characterization of p110 / mice demonstrated abnormalities in thymocyte development, T-cell activation, neutrophil migration, and oxidative burst in response to GPCR agonists (19). These mice also exhibit increased cardiac contractility (14), protection from isoproteranol-induced cardiac hypertrophy (20), and decreased leukotreine B 4 -induced degranulation of peritoneal granulocytes (21). In insulinsecreting cells, PI3 kinase has been implicated in the ability of GLP-1 to stimulate PDX-1 binding activity and DNA synthesis (2), proliferation (13), and to suppress apoptosis (22); and nonselective PI3 kinase inhibition enhances insulin secretion (7, 8). A role for the p110 isoform of PI3 kinase in islet function and development was previously unknown. One report suggests that p110 is not expressed in islets (12), whereas another demonstrates p110 activity in insulinoma cells (11). The present study identifies p110 expression in mouse and human islets and is the first to characterize glucose homeostasis in mice lacking p110. Although the insulin response of p110 / mice is severely blunted, the lack of massive glucose intolerance and diabetes may be explained by an increased responsiveness to insulin. A similar effect on ip insulin tolerance is observed in p85 / mice (23), p50 /p55 / mice (24), or mice heterozygous for Pik3r1 that encodes p85, p50, and p55 (25). However, there is no evidence for a -cell defect in those animals. In fact, whereas islet PI3 kinase activity is reduced by 80% in the p85 / mice, insulin secretion is increased and pancreatic insulin content is reduced (9), suggesting that the type 1A and type 1B PI3 kinases play distinct roles in islet function. The observation of -cell hypertrophy in the p110 / mice may suggest defective insulin processing leading to the secretion of undetected proinsulin split products rather than defective secretion per se. However, two points argue for a true secretion defect: 1) although in vivo and ex vivo insulin responses were reduced in the p110 / mice, their total pancreatic insulin content assayed with the same antibody as the in situ measurements was increased; and 2) the ex vivo insulin secretion response to arginine was not different between the wild-type and p110 / mice.

4082 Endocrinology, September 2004, 145(9):4078 4083 MacDonald et al. Brief Communications Insulin sensitivity of peripheral tissues is up-regulated in the p85 /, p50 /p55 /, or Pik3r1 / mice (23 25). Whereas the present results demonstrate intact insulin-stimulated Akt phosphorylation in p110 / heart, more direct measurements will be required to truly assess peripheral insulin sensitivity in the p110 / mice. Impaired counterregulation may contribute to the failure of plasma glucose to return to basal levels after ip insulin injection. Defective secretion of counterregulatory hormones cannot account for this, however, because fasting plasma catecholamine levels were normal and the plasma glucagon response was exaggerated at 45 min. Although the initial depression of glucagon in the p110 / mice may delay the return of blood glucose to basal levels, this should be rapidly corrected by the increased glucagon at 45 min because glucagon stimulates hepatic glucose production with a time constant of approximately 4 min (26). It therefore seems possible that the p110 / mice do not respond to these hormones appropriately, for example by initiating hepatic glucose production in response to glucagon and epinephrine. PI3 kinase mediates the proliferative and antiapoptotic effects of the G protein-coupled GLP-1 receptor (13). However, -cell mass was increased and -cell density was unchanged in the p110 / mice. Although this suggests that the G activated p110 isoform of PI3 kinase is not a positive regulator of islet growth, further studies are required to determine whether p110 mediates -cell mass expansion in response to exogenous GLP-1, and whether other PI3 kinase isoforms may be activated directly by the GLP-1 receptor. We also cannot rule out indirect G protein activation of other PI3 kinase isoforms because recent evidence suggests that GLP-1 activates type 1A PI3 kinases that are not commonly G protein linked through the trans-activation of epidermal growth factor tyrosine kinase receptors (18, 27). Although the GLP-1 receptor agonist exendin 4 was found to increase glucose tolerance in the p110 / mice, further studies are required to determine whether a more subtle phenotype is present, such as a shift in the GLP-1 dose response or altered responses to other GPCR agonists. Although not essential for the development of -cell mass or the hypoglycemic action of exendin 4, p110 is integral for appropriate -cell responsiveness to glucose. This represents a surprising finding given the presumed role of G proteinlinked PI3 kinase activity in islet growth and the ability of PI3 kinase inhibitors to stimulate insulin secretion. The p110 isoform of PI3 kinase can activate numerous downstream targets implicated as regulators of insulin secretion. These include protein kinase C, nitric oxide synthase and, interestingly, guanine nucleotide exchange factors that regulate small G proteins such as Rac and Cdc42 (10). It is also possible that defective insulin secretion in the p110 / mice results from altered -cell development due to the lack of this PI3 kinase isoform from birth. Clearly, further studies of the metabolic and electrical function of isolated p110 / -cells, and study of the acute effects of p110, are required to elucidate the nature of defective insulin secretion in these animals. Acknowledgments The authors thank Dr. Joseph Penninger (Toronto, Canada) for the p110 / mice, and Dr. Jonathan Lakey (Edmonton, Canada) and the JDRF Human Islet Distribution Program at the University of Alberta for human islets. Also, we thank Dr. Albert Salehi (Lund, Sweden) for critical reading of the manuscript. Received January 12, 2004. Accepted June 22, 2004. Address all correspondence and requests for reprints to: Dr. P. E. MacDonald, Lund University, Department of Molecular and Cellular Physiology, Tornavägen 10, BMC B11, 221 84 Lund, Sweden. E-mail: patrick.macdonald@mphy.lu.se. J.W.J. and D.Y. contributed equally and their names appear in alphabetical order. This work was supported by a grant from the Canadian Institutes of Health Research (CIHR, MOP-12898). D.Y. and Z.A. are supported by Banting and Best Diabetes Centre studentships. J.W.J. is supported by a CIHR Doctoral Studentship. P.E.M.is a CIHR Fellow, P.H.B. is a Career Investigator of the Heart and Stroke Foundation of Ontario, and M.B.W. is a CIHR Investigator. References 1. Kahn SE 2001 Clinical review 135: the importance of -cell failure in the development and progression of type 2 diabetes. J Clin Endocrinol Metab 86:4047 4058 2. Buteau J, Roduit R, Susini S, Prentki M 1999 Glucagon-like peptide-1 promotes DNA synthesis, activates phosphatidylinositol 3-kinase and increases transcription factor pancreatic and duodenal homeobox gene 1 (PDX-1) DNA binding activity in (INS-1)-cells. Diabetologia 42:856 864 3. Xu G, Stoffers DA, Habener JF, Bonner-Weir S 1999 Exendin-4 stimulates both -cell replication and neogenesis, resulting in increased -cell mass and improved glucose tolerance in diabetic rats. Diabetes 48:2270 2276 4. Farilla L, Bulotta A, Hirshberg B, Li CS, Khoury N, Noushmehr H, Bertolotto C, Di Mario U, Harlan DM, Perfetti R 2003 GLP-1 inhibits cell apoptosis and improves glucose responsiveness of freshly isolated human islets. Endocrinology 144:5149 5158 5. Li Y, Hansotia T, Yusta B, Ris F, Halban PA, Drucker DJ 2003 Glucagonlike peptide-1 receptor signaling modulates -cell apoptosis. J Biol Chem 278:471 478 6. Trumper K, Trumper A, Trusheim H, Arnold R, Goke B, Horsch D 2000 Integrative mitogenic role of protein kinase B/Akt in -cells. Ann NY Acad Sci 921:242 250 7. Zawalich WS, Zawalich KC 2000 A link between insulin resistance and hyperinsulinemia: inhibitors of phosphatidylinositol 3-kinase augment glucoseinduced insulin secretion from islets of lean, but not obese, rats. Endocrinology 141:3287 3295 8. Hagiwara S, Sakurai T, Tashiro F, Hashimoto Y, Matsuda Y, Nonomura Y, Miyazaki J 1995 An inhibitory role for phosphatidylinositol 3-kinase in insulin secretion from pancreatic B cell line MIN6. Biochem Biophys Res Commun 214:51 59 9. Eto K, Yamashita T, Tsubamoto Y, Terauchi Y, Hirose K, Kubota N, Yamashita S, Taka J, Satoh S, Sekihara H, Tobe K, Iino M, Noda M, Kimura S, Kadowaki T 2002 Phosphatidylinositol 3-kinase suppresses glucosestimulated insulin secretion by affecting post-cytosolic [Ca 2 ] elevation signals. Diabetes 51:87 97 10. Koyasu S 2003 The role of PI3K in immune cells. Nat Immunol 4:313 319 11. Trumper A, Trumper K, Trusheim H, Arnold R, Goke B, Horsch D 2001 Glucose-dependent insulinotropic polypeptide is a growth factor for (INS-1) cells by pleiotropic signaling. Mol Endocrinol 15:1559 1570 12. Bernstein HG, Keilhoff G, Reiser M, Freese S, Wetzker R 1998 Tissue distribution and subcellular localization of a G-protein activated phosphoinositide 3-kinase. An immunohistochemical study. Cell Mol Biol (Noisy-le-grand) 44:973 983 13. Buteau J, Foisy S, Rhodes CJ, Carpenter L, Biden TJ, Prentki M 2001 Protein kinase C activation mediates glucagon-like peptide-1-induced pancreatic -cell proliferation. Diabetes 50:2237 2243 14. Crackower MA, Oudit GY, Kozieradzki I, Sarao R, Sun H, Sasaki T, Hirsch E, Suzuki A, Shioi T, Irie-Sasaki J, Sah R, Cheng HY, Rybin VO, Lembo G, Fratta L, Oliveira-dos-Santos AJ, Benovic JL, Kahn CR, Izumo S, Steinberg SF, Wymann MP, Backx PH, Penninger JM 2002 Regulation of myocardial contractility and cell size by distinct PI3K-PTEN signaling pathways. Cell 110:737 749 15. Joseph JW, Koshkin V, Zhang CY, Wang J, Lowell BB, Chan CB, Wheeler MB 2002 Uncoupling protein 2 knockout mice have enhanced insulin secretory capacity after a high-fat diet. Diabetes 51:3211 3219 16. Pederson RA, Satkunarajah M, McIntosh CH, Scrocchi LA, Flamez D, Schuit F, Drucker DJ, Wheeler MB 1998 Enhanced glucose-dependent insulinotropic

MacDonald et al. Brief Communications Endocrinology, September 2004, 145(9):4078 4083 4083 polypeptide secretion and insulinotropic action in glucagon-like peptide 1 receptor / mice. Diabetes 47:1046 1052 17. Wright PH, Makulu DR, Malaisse WJ, Roberts NM, Yu PL 1968 A method for the immunoassay of insulin. Diabetes 17:537 546 18. MacDonald PE, Wang X, Xia F, El Kholy W, Targonsky E, Tsushima RG, Wheeler MB 2003 Antagonism of rat -cell voltage-dependent K currents by exendin-4 requires dual activation of the camp/pka and PI3 kinase signalling pathways. J Biol Chem 278:52446 52453 19. Sasaki T, Irie-Sasaki J, Jones RG, Oliveira-dos-Santos AJ, Stanford WL, Bolon B, Wakeham A, Itie A, Bouchard D, Kozieradzki I, Joza N, Mak TW, Ohashi PS, Suzuki A, Penninger JM 2000 Function of PI3K in thymocyte development, T cell activation, and neutrophil migration. Science 287:1040 1046 20. Oudit GY, Crackower MA, Eriksson U, Sarao R, Kozieradzki I, Sasaki T, Irie-Sasaki J, Gidrewicz D, Rybin VO, Wada T, Steinberg SF, Backx PH, Penninger JM 2003 Phosphoinositide 3-kinase -deficient mice are protected from isoproterenol-induced heart failure. Circulation 108:2147 2152 21. Ito N, Yokomizo T, Sasaki T, Kurosu H, Penninger JM, Kanaho Y, Katada T, Hanaoka K, Shimizu T 2002 Requirement of phosphatidylinositol 3-kinase activation and calcium influx for leukotriene B 4 -induced enzyme release. J Biol Chem 277:44898 44904 22. Hui H, Nourparvar A, Zhao X, Perfetti R 2003 Glucagon-like peptide-1 inhibits apoptosis of insulin-secreting cells via a cyclic 5 -adenosine monophosphatedependent protein kinase A- and a phosphatidylinositol 3-kinase-dependent pathway. Endocrinology 144:1444 1455 23. Terauchi Y, Tsuji Y, Satoh S, Minoura H, Murakami K, Okuno A, Inukai K, Asano T, Kaburagi Y, Ueki K, Nakajima H, Hanafusa T, Matsuzawa Y, Sekihara H, Yin Y, Barrett JC, Oda H, Ishikawa T, Akanuma Y, Komuro I, Suzuki M, Yamamura K, Kodama T, Suzuki H, Kadowaki T 1999 Increased insulin sensitivity and hypoglycaemia in mice lacking the p85 subunit of phosphoinositide 3-kinase. Nat Genet 21:230 235 24. Chen D, Mauvais-Jarvis F, Bluher M, Fisher SJ, Jozsi A, Goodyear LJ, Ueki K, Kahn CR 2004 p50 /p55 Phosphoinositide 3-kinase knockout mice exhibit enhanced insulin sensitivity. Mol Cell Biol 24:320 329 25. Mauvais-Jarvis F, Ueki K, Fruman DA, Hirshman MF, Sakamoto K, Goodyear LJ, Iannacone M, Accili D, Cantley LC, Kahn CR 2002 Reduced expression of the murine p85 subunit of phosphoinositide 3-kinase improves insulin signaling and ameliorates diabetes. J Clin Invest 109:141 149 26. Dobbins RL, Davis SN, Neal D, Caumo A, Cobelli C, Cherrington AD 1998 Rates of glucagon activation and deactivation of hepatic glucose production in conscious dogs. Metabolism 47:135 142 27. Buteau J, Foisy S, Joly E, Prentki M 2003 Glucagon-like peptide 1 induces pancreatic -cell proliferation via transactivation of the epidermal growth factor receptor. Diabetes 52:124 132 Endocrinology is published monthly by The Endocrine Society (http://www.endo-society.org), the foremost professional society serving the endocrine community.