Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
PLoS One ; 15(3): e0224344, 2020.
Article in English | MEDLINE | ID: mdl-32176701

ABSTRACT

A key event in the development of both major forms of diabetes is the loss of functional pancreatic islet ß-cell mass. Strategies aimed at enhancing ß-cell regeneration have long been pursued, but methods for reliably inducing human ß-cell proliferation with full retention of key functions such as glucose-stimulated insulin secretion (GSIS) are still very limited. We have previously reported that overexpression of the homeobox transcription factor NKX6.1 stimulates ß-cell proliferation, while also enhancing GSIS and providing protection against ß-cell cytotoxicity through induction of the VGF prohormone. We developed an NKX6.1 pathway screen by stably transfecting 832/13 rat insulinoma cells with a VGF promoter-luciferase reporter construct, using the resultant cell line to screen a 630,000 compound chemical library. We isolated three compounds with consistent effects to stimulate human islet cell proliferation, but not expression of NKX6.1 or VGF, suggesting an alternative mechanism of action. Further studies of the most potent of these compounds, GNF-9228, revealed that it selectively activates human ß-cell relative to α-cell proliferation and has no effect on δ-cell replication. In addition, pre-treatment, but not short term exposure of human islets to GNF-9228 enhances GSIS. GNF-9228 also protects 832/13 insulinoma cells against ER stress- and inflammatory cytokine-induced cytotoxicity. GNF-9228 stimulates proliferation via a mechanism distinct from recently emergent DYRK1A inhibitors, as it is unaffected by DYRK1A overexpression and does not activate NFAT translocation. In conclusion, we have identified a small molecule with pleiotropic positive effects on islet biology, including stimulation of human ß-cell proliferation and insulin secretion, and protection against multiple agents of cytotoxic stress.


Subject(s)
Cell Proliferation/drug effects , Insulin Secretion/drug effects , Insulin-Secreting Cells/metabolism , Insulinoma/metabolism , Protein Kinase Inhibitors/pharmacology , Animals , Cell Line, Tumor , Glucagon-Secreting Cells/metabolism , Glucagon-Secreting Cells/pathology , Glucose/pharmacology , Homeodomain Proteins/metabolism , Humans , Insulin-Secreting Cells/pathology , Insulinoma/pathology , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Rats , Dyrk Kinases
3.
PLoS One ; 12(2): e0172567, 2017.
Article in English | MEDLINE | ID: mdl-28212395

ABSTRACT

Increased ß-cell death coupled with the inability to replicate existing ß-cells drives the decline in ß-cell mass observed in the progression of both major forms of diabetes. Understanding endogenous mechanisms of islet cell survival could have considerable value for the development of novel strategies to limit ß-cell loss and thereby promote ß-cell recovery. Insulinoma cells have provided useful insight into ß-cell death pathways but observations made in cell lines sometimes fail to translate to primary islets. Here, we report dramatic differences in the temporal regulation and engagement of the apoptotic program in primary rodent islets relative to the INS-1 derived 832/13 cell line. As expected, 832/13 cells rapidly induced cell stress markers in response to ER stress or DNA damage and were fully committed to apoptosis, resulting in >80% cell death within 24 h. In contrast, primary rat islets were largely refractory to cell death in response to ER stress and DNA damage, despite rapid induction of stress markers, such as XBP-1(s), CHOP, and PUMA. Gene expression profiling revealed a general suppression of pro-apoptotic machinery, such as Apaf-1 and caspase 3, and sustained levels of pro-survival factors, such as cIAP-1, cIAP-2, and XIAP, in rat islets. Furthermore, we observed sustained induction of autophagy following chronic ER stress and found that inhibition of autophagy rendered islet ß-cells highly vulnerable to ER stress-induced cell death. We propose that islet ß-cells dampen the apoptotic response to delay the onset of cell death, providing a temporal window in which autophagy can be activated to limit cellular damage and promote survival.


Subject(s)
Apoptosis/physiology , Autophagy/physiology , Islets of Langerhans/cytology , Animals , Apoptotic Protease-Activating Factor 1 , Caspase 3/metabolism , Cell Line , Cell Survival/physiology , Cells, Cultured , Endoplasmic Reticulum Stress , Glucose/metabolism , Inhibitor of Apoptosis Proteins/metabolism , Insulin/metabolism , Insulin Secretion , Insulinoma/pathology , Islets of Langerhans/physiology , Pancreatic Neoplasms/pathology , Rats
4.
Mol Cell Biol ; 36(23): 2918-2930, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27620967

ABSTRACT

The homeodomain transcription factor Pdx-1 has important roles in pancreas and islet development as well as in ß-cell function and survival. We previously reported that Pdx-1 overexpression stimulates islet cell proliferation, but the mechanism remains unclear. Here, we demonstrate that overexpression of Pdx-1 triggers proliferation largely by a non-cell-autonomous mechanism mediated by soluble factors. Consistent with this idea, overexpression of Pdx-1 under the control of a ß-cell-specific promoter (rat insulin promoter [RIP]) stimulates proliferation of both α and ß cells, and overexpression of Pdx-1 in islets separated by a Transwell membrane from islets lacking Pdx-1 overexpression activates proliferation in the untreated islets. Microarray and gene ontology (GO) analysis identified inhibin beta-B (Inhbb), an activin subunit and member of the transforming growth factor ß (TGF-ß) superfamily, as a Pdx-1-responsive gene. Overexpression of Inhbb or addition of activin B stimulates rat islet cell and ß-cell proliferation, and the activin receptors RIIA and RIIB are required for the full proliferative effects of Pdx-1 in rat islets. In human islets, Inhbb overexpression stimulates total islet cell proliferation and potentiates Pdx-1-stimulated proliferation of total islet cells and ß cells. In sum, this study identifies a mechanism by which Pdx-1 induces a soluble factor that is sufficient to stimulate both rat and human islet cell proliferation.


Subject(s)
Homeodomain Proteins/metabolism , Inhibin-beta Subunits/genetics , Insulin/genetics , Islets of Langerhans/cytology , Trans-Activators/metabolism , Animals , Cell Proliferation , Cells, Cultured , Gene Expression Profiling , Glucagon-Secreting Cells/cytology , Glucagon-Secreting Cells/metabolism , Humans , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Islets of Langerhans/metabolism , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic , Rats
5.
Mol Endocrinol ; 29(9): 1243-53, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26218441

ABSTRACT

Better understanding how glucagon-like peptide 1 (GLP-1) promotes pancreatic ß-cell function and/or mass may uncover new treatment for type 2 diabetes. In this study, we investigated the potential involvement of microRNAs (miRNAs) in the effect of GLP-1 on glucose-stimulated insulin secretion. miRNA levels in INS-1 cells and isolated rodent and human islets treated with GLP-1 in vitro and in vivo (with osmotic pumps) were measured by real-time quantitative PCR. The role of miRNAs on insulin secretion was studied by transfecting INS-1 cells with either precursors or antisense inhibitors of miRNAs. Among the 250 miRNAs surveyed, miR-132 and miR-212 were significantly up-regulated by GLP-1 by greater than 2-fold in INS-1 832/3 cells, which were subsequently reproduced in freshly isolated rat, mouse, and human islets, as well as the islets from GLP-1 infusion in vivo in mice. The inductions of miR-132 and miR-212 by GLP-1 were correlated with cAMP production and were blocked by the protein kinase A inhibitor H-89 but not affected by the exchange protein activated by cAMP activator 8-pCPT-2'-O-Me-cAMP-AM. GLP-1 failed to increase miR-132 or miR-212 expression levels in the 832/13 line of INS-1 cells, which lacks robust cAMP and insulin responses to GLP-1 treatment. Overexpression of miR-132 or miR-212 significantly enhanced glucose-stimulated insulin secretion in both 832/3 and 832/13 cells, and restored insulin responses to GLP-1 in INS-1 832/13 cells. GLP-1 increases the expression of miRNAs 132 and 212 via a cAMP/protein kinase A-dependent pathway in pancreatic ß-cells. Overexpression of miR-132 or miR-212 enhances glucose and GLP-1-stimulated insulin secretion.


Subject(s)
Glucagon-Like Peptide 1/pharmacology , Insulin-Secreting Cells/metabolism , MicroRNAs/biosynthesis , Animals , Cell Line, Tumor , Cyclic AMP/analogs & derivatives , Cyclic AMP/biosynthesis , Cyclic AMP/genetics , Cyclic AMP/pharmacology , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Diabetes Mellitus, Type 2/metabolism , Glucose/metabolism , Humans , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/cytology , Isoquinolines/pharmacology , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Protein Kinase Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , Sulfonamides/pharmacology
6.
Proc Natl Acad Sci U S A ; 111(14): 5242-7, 2014 Apr 08.
Article in English | MEDLINE | ID: mdl-24706823

ABSTRACT

Loss of functional ß-cell mass is a hallmark of type 1 and type 2 diabetes, and methods for restoring these cells are needed. We have previously reported that overexpression of the homeodomain transcription factor NK6 homeobox 1 (Nkx6.1) in rat pancreatic islets induces ß-cell proliferation and enhances glucose-stimulated insulin secretion, but the pathway by which Nkx6.1 activates ß-cell expansion has not been defined. Here, we demonstrate that Nkx6.1 induces expression of the nuclear receptor subfamily 4, group A, members 1 and 3 (Nr4a1 and Nr4a3) orphan nuclear receptors, and that these factors are both necessary and sufficient for Nkx6.1-mediated ß-cell proliferation. Consistent with this finding, global knockout of Nr4a1 results in a decrease in ß-cell area in neonatal and young mice. Overexpression of Nkx6.1 and the Nr4a receptors results in increased expression of key cell cycle inducers E2F transcription factor 1 and cyclin E1. Furthermore, Nkx6.1 and Nr4a receptors induce components of the anaphase-promoting complex, including ubiquitin-conjugating enzyme E2C, resulting in degradation of the cell cycle inhibitor p21. These studies identify a unique bipartite pathway for activation of ß-cell proliferation, suggesting several unique targets for expansion of functional ß-cell mass.


Subject(s)
Cell Proliferation , DNA-Binding Proteins/physiology , Homeodomain Proteins/physiology , Islets of Langerhans/cytology , Nerve Tissue Proteins/physiology , Nuclear Receptor Subfamily 4, Group A, Member 1/physiology , Animals , Animals, Newborn , Chromatin Immunoprecipitation , Homeodomain Proteins/genetics , Male , Mice, Knockout , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , Rats , Rats, Wistar , Ubiquitin-Conjugating Enzymes/metabolism , Up-Regulation
7.
Mol Cell Biol ; 33(20): 4017-29, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23938296

ABSTRACT

The homeodomain transcription factor Pdx-1 has important roles in pancreatic development and ß-cell function and survival. In the present study, we demonstrate that adenovirus-mediated overexpression of Pdx-1 in rat or human islets also stimulates cell replication. Moreover, cooverexpression of Pdx-1 with another homeodomain transcription factor, Nkx6.1, has an additive effect on proliferation compared to either factor alone, implying discrete activating mechanisms. Consistent with this, Nkx6.1 stimulates mainly ß-cell proliferation, whereas Pdx-1 stimulates both α- and ß-cell proliferation. Furthermore, cyclins D1/D2 are upregulated by Pdx-1 but not by Nkx6.1, and inhibition of cdk4 blocks Pdx-1-stimulated but not Nkx6.1-stimulated islet cell proliferation. Genes regulated by Pdx-1 but not Nkx6.1 were identified by microarray analysis. Two members of the transient receptor potential cation (TRPC) channel family, TRPC3 and TRPC6, are upregulated by Pdx-1 overexpression, and small interfering RNA (siRNA)-mediated knockdown of TRPC3/6 or TRPC6 alone inhibits Pdx-1-induced but not Nkx6.1-induced islet cell proliferation. Pdx-1 also stimulates extracellular signal-regulated kinase 1 and 2 (ERK1/2) phosphorylation, an effect partially blocked by knockdown of TRPC3/6, and blockade of ERK1/2 activation with a MEK1/2 inhibitor partially impairs Pdx-1-stimulated proliferation. These studies define a pathway by which overexpression of Pdx-1 activates islet cell proliferation that is distinct from and additive to a pathway activated by Nkx6.1.


Subject(s)
Glucagon-Secreting Cells/metabolism , Homeodomain Proteins/genetics , Insulin-Secreting Cells/metabolism , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/genetics , TRPC Cation Channels/genetics , Trans-Activators/genetics , Adenoviridae/genetics , Animals , Cell Proliferation , Gene Expression Regulation , Genes, Reporter , Genetic Vectors , Glucagon-Secreting Cells/cytology , Homeodomain Proteins/metabolism , Humans , Insulin-Secreting Cells/cytology , Male , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Rats , Rats, Wistar , Signal Transduction , TRPC Cation Channels/metabolism , TRPC6 Cation Channel , Trans-Activators/metabolism
8.
J Biol Chem ; 288(32): 23128-40, 2013 Aug 09.
Article in English | MEDLINE | ID: mdl-23788641

ABSTRACT

Recent studies have shown that the pyruvate-isocitrate cycling pathway, involving the mitochondrial citrate/isocitrate carrier and the cytosolic NADP-dependent isocitrate dehydrogenase (ICDc), is involved in control of glucose-stimulated insulin secretion (GSIS). Here we demonstrate that pyruvate-isocitrate cycling regulates expression of the voltage-gated potassium channel family member Kv2.2 in islet ß-cells. siRNA-mediated suppression of ICDc, citrate/isocitrate carrier, or Kv2.2 expression impaired GSIS, and the effect of ICDc knockdown was rescued by re-expression of Kv2.2. Moreover, chronic exposure of ß-cells to elevated fatty acids, which impairs GSIS, resulted in decreased expression of Kv2.2. Surprisingly, knockdown of ICDc or Kv2.2 increased rather than decreased outward K(+) current in the 832/13 ß-cell line. Immunoprecipitation studies demonstrated interaction of Kv2.1 and Kv2.2, and co-overexpression of the two channels reduced outward K(+) current compared with overexpression of Kv2.1 alone. Also, siRNA-mediated knockdown of ICDc enhanced the suppressive effect of the Kv2.1-selective inhibitor stromatoxin1 on K(+) currents. Our data support a model in which a key function of the pyruvate-isocitrate cycle is to maintain levels of Kv2.2 expression sufficient to allow it to serve as a negative regulator of Kv channel activity.


Subject(s)
Gene Expression Regulation/physiology , Glucose/metabolism , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Isocitrates/metabolism , Pyruvic Acid/metabolism , Shab Potassium Channels/biosynthesis , Animals , Gene Expression Regulation/drug effects , Glucose/genetics , Insulin Secretion , Insulin-Secreting Cells/cytology , Ion Channel Gating/drug effects , Ion Channel Gating/physiology , Ion Transport/drug effects , Ion Transport/physiology , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Male , Models, Biological , Peptides/pharmacology , Potassium/metabolism , Rats , Rats, Sprague-Dawley , Shab Potassium Channels/antagonists & inhibitors , Shab Potassium Channels/genetics , Spider Venoms/pharmacology
9.
J Pharmacol Exp Ther ; 344(2): 407-16, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23161216

ABSTRACT

The voltage-gated potassium channels Kv2.1 and Kv2.2 are highly expressed in pancreatic islets, yet their contribution to islet hormone secretion is not fully understood. Here we investigate the role of Kv2 channels in pancreatic islets using a combination of genetic and pharmacologic approaches. Pancreatic ß-cells from Kv2.1(-/-) mice possess reduced Kv current and display greater glucose-stimulated insulin secretion (GSIS) relative to WT ß-cells. Inhibition of Kv2.x channels with selective peptidyl [guangxitoxin-1E (GxTX-1E)] or small molecule (RY796) inhibitors enhances GSIS in isolated wild-type (WT) mouse and human islets, but not in islets from Kv2.1(-/-) mice. However, in WT mice neither inhibitor improved glucose tolerance in vivo. GxTX-1E and RY796 enhanced somatostatin release in isolated human and mouse islets and in situ perfused pancreata from WT and Kv2.1(-/-) mice. Kv2.2 silencing in mouse islets by adenovirus-small hairpin RNA (shRNA) specifically enhanced islet somatostatin, but not insulin, secretion. In mice lacking somatostatin receptor 5, GxTX-1E stimulated insulin secretion and improved glucose tolerance. Collectively, these data show that Kv2.1 regulates insulin secretion in ß-cells and Kv2.2 modulates somatostatin release in δ-cells. Development of selective Kv2.1 inhibitors without cross inhibition of Kv2.2 may provide new avenues to promote GSIS for the treatment of type 2 diabetes.


Subject(s)
Insulin-Secreting Cells/metabolism , Insulin/metabolism , Shab Potassium Channels/metabolism , Somatostatin/metabolism , Adult , Animals , Arthropod Proteins , Benzamides/pharmacology , Cells, Cultured , Electrophysiological Phenomena , Female , Glucose/pharmacology , Humans , Insulin Secretion , Insulin-Secreting Cells/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Patch-Clamp Techniques , Peptides/pharmacology , Potassium Channel Blockers/pharmacology , Protein Binding , Receptors, Somatostatin/genetics , Receptors, Somatostatin/metabolism , Shab Potassium Channels/antagonists & inhibitors , Shab Potassium Channels/genetics , Spider Venoms/pharmacology , Young Adult
10.
Cell Metab ; 16(1): 33-43, 2012 Jul 03.
Article in English | MEDLINE | ID: mdl-22768837

ABSTRACT

Deterioration of functional islet ß-cell mass is the final step in progression to Type 2 diabetes. We previously reported that overexpression of Nkx6.1 in rat islets has the dual effects of enhancing glucose-stimulated insulin secretion (GSIS) and increasing ß-cell replication. Here we show that Nkx6.1 strongly upregulates the prohormone VGF in rat islets and that VGF is both necessary and sufficient for Nkx6.1-mediated enhancement of GSIS. Moreover, the VGF-derived peptide TLQP-21 potentiates GSIS in rat and human islets and improves glucose tolerance in vivo. Chronic injection of TLQP-21 in prediabetic ZDF rats preserves islet mass and slows diabetes onset. TLQP-21 prevents islet cell apoptosis by a pathway similar to that used by GLP-1, but independent of the GLP-1, GIP, or VIP receptors. Unlike GLP-1, TLQP-21 does not inhibit gastric emptying or increase heart rate. We conclude that TLQP-21 is a targeted agent for enhancing islet ß-cell survival and function.


Subject(s)
Cell Survival/drug effects , Diabetes Mellitus, Type 2/prevention & control , Hypoglycemic Agents/pharmacology , Insulin-Secreting Cells/physiology , Peptide Fragments/pharmacology , Animals , Apoptosis/drug effects , Area Under Curve , Blood Glucose , Cells, Cultured , Cyclic AMP/metabolism , Diabetes Mellitus, Type 2/pathology , Gastric Emptying/drug effects , Gene Expression , Glucose/physiology , Heart Rate/drug effects , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Hyperglycemia/prevention & control , Hypoglycemic Agents/therapeutic use , Insulin/blood , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Male , Neuropeptides/genetics , Neuropeptides/metabolism , Peptide Fragments/physiology , Peptide Fragments/therapeutic use , Rats , Rats, Wistar , Trans-Activators/genetics , Trans-Activators/metabolism
11.
PLoS One ; 6(4): e18517, 2011 Apr 06.
Article in English | MEDLINE | ID: mdl-21494687

ABSTRACT

UFM1 is a member of the ubiquitin like protein family. While the enzymatic cascade of UFM1 conjugation has been elucidated in recent years, the biological function remains largely unknown. In this report we demonstrate that the recently identified C20orf116, which we name UFM1-binding protein 1 containing a PCI domain (UFBP1), and CDK5RAP3 interact with UFM1. Components of the UFM1 conjugation pathway (UFM1, UFBP1, UFL1 and CDK5RAP3) are highly expressed in pancreatic islets of Langerhans and some other secretory tissues. Co-localization of UFM1 with UFBP1 in the endoplasmic reticulum (ER) depends on UFBP1. We demonstrate that ER stress, which is common in secretory cells, induces expression of Ufm1, Ufbp1 and Ufl1 in the beta-cell line INS-1E. siRNA-mediated Ufm1 or Ufbp1 knockdown enhances apoptosis upon ER stress. Silencing the E3 enzyme UFL1, results in similar outcomes, suggesting that UFM1-UFBP1 conjugation is required to prevent ER stress-induced apoptosis. Together, our data suggest that UFM1-UFBP1 participate in preventing ER stress-induced apoptosis in protein secretory cells.


Subject(s)
Apoptosis , Carrier Proteins/metabolism , Cytoprotection , Endoplasmic Reticulum/pathology , Insulin-Secreting Cells/cytology , Proteins/metabolism , Stress, Physiological , Amino Acid Sequence , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cytoprotection/drug effects , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Gene Expression Profiling , Gene Expression Regulation/drug effects , Gene Knockdown Techniques , Glucose/pharmacology , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Male , Mass Spectrometry , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Nerve Tissue Proteins/metabolism , Protein Binding/drug effects , Protein Transport/drug effects , Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Stress, Physiological/drug effects , Stress, Physiological/genetics
12.
Sci Signal ; 3(113): ra19, 2010 Mar 16.
Article in English | MEDLINE | ID: mdl-20234002

ABSTRACT

Parasympathetic stimulation of pancreatic islets augments glucose-stimulated insulin secretion by inducing inositol trisphosphate receptor (IP(3)R)-mediated calcium ion (Ca2+) release. Ankyrin-B binds to the IP(3)R and is enriched in pancreatic beta cells. We found that ankyrin-B-deficient islets displayed impaired potentiation of insulin secretion by the muscarinic agonist carbachol, blunted carbachol-mediated intracellular Ca2+ release, and reduced the abundance of IP3R. Ankyrin-B-haploinsufficient mice exhibited hyperglycemia after oral ingestion but not after intraperitoneal injection of glucose, consistent with impaired parasympathetic potentiation of glucose-stimulated insulin secretion. The R1788W mutation of ankyrin-B impaired its function in pancreatic islets and is associated with type 2 diabetes in Caucasians and Hispanics. Thus, defective glycemic regulation through loss of ankyrin-B-dependent stabilization of IP3R is a potential risk factor for type 2 diabetes.


Subject(s)
Ankyrins/metabolism , Diabetes Mellitus, Type 2/metabolism , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Parasympathetic Nervous System/metabolism , Animals , Ankyrins/deficiency , Ankyrins/genetics , Calcium/metabolism , Carbachol/metabolism , Glucose/metabolism , Immunoblotting , Insulin Secretion , Mice , Microscopy, Fluorescence , Mutation, Missense , Polymorphism, Single Nucleotide/genetics , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , Risk Factors
13.
Mol Endocrinol ; 23(6): 881-92, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19246514

ABSTRACT

Collectrin is a downstream target of the transcription factor hepatocyte nuclear factor-1alpha (HNF-1alpha), which is mutated in maturity-onset diabetes of the young subtype 3 (MODY3). Evidence from transgenic mouse models with collectrin overexpression in pancreatic islets suggests divergent roles for collectrin in influencing beta-cell mass and insulin exocytosis. To clarify the function of collectrin in the pancreas, we used a mouse line with targeted deletion of the gene. We examined pancreas morphology, glucose homeostasis by ip glucose tolerance testing (IPGTT) and insulin tolerance testing (IPITT), and pancreas function by in vivo acute-phase insulin response determination and glucose-stimulated insulin secretion from isolated islets. We find no difference in either pancreas morphology or function between wild-type and collectrin-deficient animals (Tmem27(-/y)). However, we note that by 6 months of age, Tmem27(-/y) mice exhibit increased insulin sensitivity by IPITT and decreased adiposity by dual-energy x-ray absorptiometry scanning compared with wild-type. We have previously reported that Tmem27(-/y) mice exhibit profound aminoaciduria due to failed renal recovery. We now demonstrate that Tmem27(-/y) animals also display inappropriate excretion of some short-chain acylcarnitines derived from amino acid and fatty acid oxidation. We provide further evidence for compensatory up-regulation of oxidative metabolism in Tmem27(-/y) mice, along with enhanced protein turnover associated with preserved lean mass even out to 1.5 yr of age. Our studies suggest that collectrin-deficient mice activate a number of adaptive mechanisms to defend energy homeostasis in the setting of ongoing nutrient losses.


Subject(s)
Hepatocyte Nuclear Factor 1-alpha/metabolism , Insulin/metabolism , Membrane Glycoproteins/deficiency , Acute-Phase Reaction/metabolism , Adiposity/drug effects , Amino Acids/blood , Animals , Body Weight/drug effects , Cell Proliferation/drug effects , Fasting/blood , Food , Glucose/pharmacology , Glucose Tolerance Test , Insulin Resistance , Insulin Secretion , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Membrane Glycoproteins/metabolism , Mice , Phenotype , Protein Processing, Post-Translational/drug effects , Thinness/blood , Thinness/metabolism , Time Factors
14.
Dig Dis Sci ; 54(3): 481-7, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18618254

ABSTRACT

BACKGROUND: Agenesis of the dorsal pancreas is a very rare congenital pancreatic malformation and is associated with some other diseases. METHODS: A PubMed search revealed 53 cases of agenesis of the dorsal pancreas. RESULTS: In 28 patients with this congenital malformation hyperglycemia was demonstrated, 27 had abdominal pain, 16 had pancreatitis, 14 had an enlarged or prominent pancreatic head visible on computed tomography, and in a few cases, polysplenia, which may occur with various congenital anomalies of visceral organs, was described. CONCLUSIONS: Difficulties involved in obtaining a firm diagnosis have led to a variety of terms being used to describe this congenital disease. Diagnosis of agenesis of the dorsal pancreas is inconclusive without demonstration of the absence of the dorsal pancreatic duct. Here we describe the embryological development of the pancreas, the so-far known cases of agenesis of the dorsal pancreas with associated medical problems, and the diagnostic measures to find the right conclusions.


Subject(s)
Diabetes Mellitus/congenital , Pancreas/abnormalities , Pancreatic Diseases/congenital , Humans , Pancreatic Diseases/complications
15.
J Biol Chem ; 283(43): 28909-17, 2008 Oct 24.
Article in English | MEDLINE | ID: mdl-18755687

ABSTRACT

We have previously demonstrated a role for pyruvate cycling in glucose-stimulated insulin secretion (GSIS). Some of the possible pyruvate cycling pathways are completed by conversion of malate to pyruvate by malic enzyme. Using INS-1-derived 832/13 cells, it has recently been shown by other laboratories that NADP-dependent cytosolic malic enzyme (MEc), but not NAD-dependent mitochondrial malic enzyme (MEm), regulates GSIS. In the current study, we show that small interfering RNA-mediated suppression of either MEm or MEc results in decreased GSIS in both 832/13 cells and a new and more glucose- and incretin-responsive INS-1-derived cell line, 832/3. The effect of MEm to suppress GSIS in these cell lines was linked to a substantial decrease in cell growth, whereas MEc suppression resulted in decreased NADPH, shown previously to be correlated with GSIS. However, adenovirus-mediated delivery of small interfering RNAs specific to MEc and MEm to isolated rat islets, while leading to effective suppression of the targets transcripts, had no effect on GSIS. Furthermore, islets isolated from MEc-null MOD1(-/-) mice exhibit normal glucose- and potassium-stimulated insulin secretion. These results indicate that pyruvate-malate cycling does not control GSIS in primary rodent islets.


Subject(s)
Cytosol/metabolism , Glucose/chemistry , Insulin/metabolism , Islets of Langerhans/metabolism , Malate Dehydrogenase/chemistry , Mitochondria/metabolism , Animals , Gene Silencing , Insulin Secretion , Male , Mice , Models, Biological , Protein Isoforms , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley
16.
Mol Cell Biol ; 28(10): 3465-76, 2008 May.
Article in English | MEDLINE | ID: mdl-18347054

ABSTRACT

The homeodomain transcription factor Nkx6.1 plays an important role in pancreatic islet beta-cell development, but its effects on adult beta-cell function, survival, and proliferation are not well understood. In the present study, we demonstrated that treatment of primary rat pancreatic islets with a cytomegalovirus promoter-driven recombinant adenovirus containing the Nkx6.1 cDNA (AdCMV-Nkx6.1) causes dramatic increases in [methyl-(3)H] thymidine and 5-bromo-2'-deoxyuridine (BrdU) incorporation and in the number of cells per islet relative to islets treated with a control adenovirus (AdCMV-betaGAL), whereas suppression of Nkx6.1 expression reduces thymidine incorporation. Immunocytochemical studies reveal that >80% of BrdU-positive cells in AdCMV-Nkx6.1-treated islets are beta cells. Microarray, real-time PCR, and immunoblot analyses reveal that overexpression of Nkx6.1 in rat islets causes concerted upregulation of a cadre of cell cycle control genes, including those encoding cyclins A, B, and E, and several regulatory kinases. Cyclin E is upregulated earlier than the other cyclins, and adenovirus-mediated overexpression of cyclin E is shown to be sufficient to activate islet cell proliferation. Moreover, chromatin immunoprecipitation assays demonstrate direct interaction of Nkx6.1 with the cyclin A2 and B1 genes. Overexpression of Nkx6.1 in rat islets caused a clear enhancement of glucose-stimulated insulin secretion (GSIS), whereas overexpression of Nkx6.1 in human islets caused an increase in the level of [(3)H]thymidine incorporation that was twice the control level, along with complete retention of GSIS. We conclude that Nkx6.1 is among the very rare factors capable of stimulating beta-cell replication with retention or enhancement of function, properties that may be exploitable for expansion of beta-cell mass in treatment of both major forms of diabetes.


Subject(s)
Homeodomain Proteins/metabolism , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Adenoviridae/genetics , Animals , Base Sequence , Cell Cycle/genetics , Cell Proliferation , Cyclin A/genetics , Cyclin A2 , Cyclin B/genetics , Cyclin B1 , Cyclins/metabolism , DNA Primers/genetics , Gene Expression , Glucose/pharmacology , Homeodomain Proteins/antagonists & inhibitors , Homeodomain Proteins/genetics , Humans , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/drug effects , Kinetics , Models, Biological , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Rats , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Tissue Culture Techniques
17.
Mol Endocrinol ; 22(5): 1251-9, 2008 May.
Article in English | MEDLINE | ID: mdl-18258687

ABSTRACT

Both major forms of diabetes involve a decline in beta-cell mass, mediated by autoimmune destruction of insulin-producing cells in type 1 diabetes and by increased rates of apoptosis secondary to metabolic stress in type 2 diabetes. Methods for controlled expansion of beta-cell mass are currently not available but would have great potential utility for treatment of these diseases. In the current study, we demonstrate that overexpression of trefoil factor 3 (TFF3) in rat pancreatic islets results in a 4- to 5-fold increase in [(3)H]thymidine incorporation, with full retention of glucose-stimulated insulin secretion. This increase was almost exclusively due to stimulation of beta-cell replication, as demonstrated by studies of bromodeoxyuridine incorporation and co-immunofluorescence analysis with anti-bromodeoxyuridine and antiinsulin or antiglucagon antibodies. The proliferative effect of TFF3 required the presence of serum or 0.5 ng/ml epidermal growth factor. The ability of TFF3 overexpression to stimulate proliferation of rat islets in serum was abolished by the addition of epidermal growth factor receptor antagonist AG1478. Furthermore, TFF3-induced increases in [3H]thymidine incorporation in rat islets cultured in serum was blocked by overexpression of a dominant-negative Akt protein or treatment with triciribine, an Akt inhibitor. Finally, overexpression of TFF3 also caused a doubling of [3H]thymidine incorporation in human islets. In summary, our findings reveal a novel TFF3-mediated pathway for stimulation of beta-cell replication that could ultimately be exploited for expansion or preservation of islet beta-cell mass.


Subject(s)
Insulin-Secreting Cells/metabolism , Islets of Langerhans/metabolism , Neuropeptides/physiology , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cells, Cultured , Epidermal Growth Factor/pharmacology , Humans , Immunoblotting , Insulin-Secreting Cells/cytology , Islets of Langerhans/cytology , Male , Neuropeptides/genetics , Neuropeptides/metabolism , Oncogene Protein v-akt/metabolism , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Thymidine/metabolism , Transfection , Trefoil Factor-3
18.
Mol Endocrinol ; 21(3): 765-73, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17185391

ABSTRACT

Recent advances in functional genomics afford the opportunity to interrogate the expression profiles of thousands of genes simultaneously and examine the function of these genes in a high-throughput manner. In this study, we describe a rational and efficient approach to identifying novel regulators of insulin secretion by the pancreatic beta-cell. Computational analysis of expression profiles of several mouse and cellular models of impaired insulin secretion identified 373 candidate genes involved in regulation of insulin secretion. Using RNA interference, we assessed the requirements of 10 of these candidates and identified four genes (40%) as being essential for normal insulin secretion. Among the genes identified was Hadhsc, which encodes short-chain 3-hydroxyacyl-coenzyme A dehydrogenase (SCHAD), an enzyme of mitochondrial beta-oxidation of fatty acids whose mutation results in congenital hyperinsulinism. RNA interference-mediated gene suppression of Hadhsc in insulinoma cells and primary rodent islets revealed enhanced basal but normal glucose-stimulated insulin secretion. This increase in basal insulin secretion was not attenuated by the opening of the KATP channel with diazoxide, suggesting that SCHAD regulates insulin secretion through a KATP channel-independent mechanism. Our results suggest a molecular explanation for the hyperinsulinemia hypoglycemic seen in patients with SCHAD deficiency.


Subject(s)
Butyryl-CoA Dehydrogenase/physiology , Genomics/methods , Insulin-Secreting Cells , Insulin/metabolism , Potassium Channels/physiology , Animals , Butyryl-CoA Dehydrogenase/genetics , Cells, Cultured , Gene Expression Profiling , Insulin Secretion , Insulin-Secreting Cells/enzymology , Insulin-Secreting Cells/metabolism , Mice , Mice, Transgenic , Models, Biological , RNA Interference , Rats
19.
Proc Natl Acad Sci U S A ; 103(22): 8469-74, 2006 May 30.
Article in English | MEDLINE | ID: mdl-16709667

ABSTRACT

This study describes a method of gene delivery to pancreatic islets of adult, living animals by ultrasound targeted microbubble destruction (UTMD). The technique involves incorporation of plasmids into the phospholipid shell of gas-filled microbubbles, which are then infused into rats and destroyed within the pancreatic microcirculation with ultrasound. Specific delivery of genes to islet beta cells by UTMD was achieved by using a plasmid containing a rat insulin 1 promoter (RIP), and reporter gene expression was regulated appropriately by glucose in animals that received a RIP-luciferase plasmid. To demonstrate biological efficacy, we used UTMD to deliver RIP-human insulin and RIP-hexokinase I plasmids to islets of adult rats. Delivery of the former plasmid resulted in clear increases in circulating human C-peptide and decreased blood glucose levels, whereas delivery of the latter plasmid resulted in a clear increase in hexokinase I protein expression in islets, increased insulin levels in blood, and decreased circulating glucose levels. We conclude that UTMD allows relatively noninvasive delivery of genes to pancreatic islets with an efficiency sufficient to modulate beta cell function in adult animals.


Subject(s)
Gene Transfer Techniques , Islets of Langerhans/metabolism , Microbubbles , Animals , Blood Glucose/metabolism , C-Peptide/blood , Gene Expression , Genes, Reporter/genetics , Hexokinase/genetics , Hexokinase/metabolism , Humans , Insulin/genetics , Insulin/metabolism , Insulin Secretion , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Microscopy, Confocal , Rats , Rats, Sprague-Dawley , Time Factors , Ultrasonics , Red Fluorescent Protein
20.
Diabetes ; 55(5): 1398-406, 2006 May.
Article in English | MEDLINE | ID: mdl-16644697

ABSTRACT

Type 1 diabetes results from islet beta-cell death and dysfunction induced by an autoimmune mechanism. Proinflammatory cytokines such as interleukin-1beta and gamma-interferon are mediators of this beta-cell cytotoxicity, but the mechanism by which damage occurs is not well understood. In the current study, we present multiple lines of evidence supporting the conclusion that cytokine-induced killing of rat beta-cells occurs predominantly by a nonapoptotic mechanism, including the following: 1) A rat beta-cell line selected for resistance to cytokine-induced cytotoxicity (833/15) is equally sensitive to killing by the apoptosis-inducing agents camptothecin and etoposide as a cytokine-sensitive cell line (832/13). 2) Overexpression of a constitutively active form of the antiapoptotic protein kinase Akt1 in 832/13 cells provides significant protection against cell killing induced by camptothecin and etoposide but no protection against cytokine-mediated damage. 3) Small interfering RNA-mediated suppression of the proapoptotic protein Bax enhances viability of 832/13 cells upon exposure to the known apoptosis-inducing drugs but not the inflammatory cytokines. 4) Exposure of primary rat islets or 832/13 cells to the inflammatory cytokines causes cell death as evidenced by the release of adenylate kinase activity into the cell medium, with no attendant increase in caspase 3 activation or annexin V staining. In contrast, camptothecin- and etoposide-induced killing is associated with robust increases in caspase 3 activation and annexin V staining. 5) Camptothecin increases cellular ATP levels, whereas inflammatory cytokines lower ATP levels in both beta-cell lines and primary islets. We conclude that proinflammatory cytokines cause beta-cell cytotoxicity primarily through a nonapoptotic mechanism linked to a decline in ATP levels.


Subject(s)
Apoptosis/physiology , Cytokines/pharmacology , Islets of Langerhans/cytology , Animals , Cell Line , Cell Survival/drug effects , Cells, Cultured , Gene Expression Regulation/drug effects , Interferon-gamma/pharmacology , Interleukin-1/pharmacology , Islets of Langerhans/drug effects , Islets of Langerhans/physiology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/genetics , Rats , Transfection , bcl-2 Homologous Antagonist-Killer Protein/genetics , bcl-2-Associated X Protein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...