151
|
Abstract
Insulin secretion is one of the most characteristic features of β-cell physiology. As it plays a central role in glucose regulation, a number of experimental and theoretical studies have been performed since the discovery of the pancreatic β-cell. This review article aims to give an overview of the mathematical approaches to insulin secretion. Beginning with the bursting electrical activity in pancreatic β-cells, we describe effects of the gap-junction coupling between β-cells on the dynamics of insulin secretion. Then, implications of paracrine interactions among such islet cells as α-, β-, and δ-cells are discussed. Finally, we present mathematical models which incorporate effects of glycolysis and mitochondrial glucose metabolism on the control of insulin secretion.
Collapse
Affiliation(s)
- Kyungreem Han
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul, South Korea
| | | | | | | |
Collapse
|
152
|
FoxO1 gain of function in the pancreas causes glucose intolerance, polycystic pancreas, and islet hypervascularization. PLoS One 2012; 7:e32249. [PMID: 22384192 PMCID: PMC3285669 DOI: 10.1371/journal.pone.0032249] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2011] [Accepted: 01/24/2012] [Indexed: 12/31/2022] Open
Abstract
Genetic studies revealed that the ablation of insulin/IGF-1 signaling in the pancreas causes diabetes. FoxO1 is a downstream transcription factor of insulin/IGF-1 signaling. We previously reported that FoxO1 haploinsufficiency restored β cell mass and rescued diabetes in IRS2 knockout mice. However, it is still unclear whether FoxO1 dysregulation in the pancreas could be the cause of diabetes. To test this hypothesis, we generated transgenic mice overexpressing constitutively active FoxO1 specifically in the pancreas (TG). TG mice had impaired glucose tolerance and some of them indeed developed diabetes due to the reduction of β cell mass, which is associated with decreased Pdx1 and MafA in β cells. We also observed increased proliferation of pancreatic duct epithelial cells in TG mice and some mice developed a polycystic pancreas as they aged. Furthermore, TG mice exhibited islet hypervascularities due to increased VEGF-A expression in β cells. We found FoxO1 binds to the VEGF-A promoter and regulates VEGF-A transcription in β cells. We propose that dysregulation of FoxO1 activity in the pancreas could account for the development of diabetes and pancreatic cysts.
Collapse
|
153
|
Pansuria M, Xi H, Li L, Yang XF, Wang H. Insulin resistance, metabolic stress, and atherosclerosis. Front Biosci (Schol Ed) 2012; 4:916-31. [PMID: 22202099 PMCID: PMC3319745 DOI: 10.2741/s308] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Atherosclerosis, a pathological process that underlies the development of cardiovascular disease, is the primary cause of morbidity and mortality in patients with type 2 diabetes mellitus (T2DM). T2DM is characterized by hyperglycemia and insulin resistance (IR), in which target tissues fail to respond to insulin. Systemic IR is associated with impaired insulin signaling in the metabolic tissues and vasculature. Insulin receptor is highly expressed in the liver, muscle, pancreas, and adipose tissue. It is also expressed in vascular cells. It has been suggested that insulin signaling in vascular cells regulates cell proliferation and vascular function. In this review, we discuss the association between IR, metabolic stress, and atherosclerosis with focus on 1) tissue and cell distribution of insulin receptor and its differential signaling transduction and 2) potential mechanism of insulin signaling impairment and its role in the development of atherosclerosis and vascular function in metabolic disorders including hyperglycemia, hypertension, dyslipidemia, and hyperhomocysteinemia. We propose that insulin signaling impairment is the foremost biochemical mechanism underlying increased cardiovascular morbidity and mortality in atherosclerosis, T2DM, and metabolic syndrome.
Collapse
Affiliation(s)
- Meghana Pansuria
- Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA, 19140
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA, 19140
| | - Hang Xi
- Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA, 19140
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA, 19140
| | - Le Li
- Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA, 19140
- School of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, PR, China
| | - Xiao-Feng Yang
- Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA, 19140
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA, 19140
| | - Hong Wang
- Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA, 19140
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA, 19140
- Thrombosis Research Center of Temple University School of Medicine, Philadelphia, PA, 19140
| |
Collapse
|
154
|
Tarabra E, Pelengaris S, Khan M. A simple matter of life and death-the trials of postnatal Beta-cell mass regulation. Int J Endocrinol 2012; 2012:516718. [PMID: 22577380 PMCID: PMC3346985 DOI: 10.1155/2012/516718] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2011] [Accepted: 12/31/2011] [Indexed: 12/17/2022] Open
Abstract
Pancreatic beta-cells, which secrete the hormone insulin, are the key arbiters of glucose homeostasis. Defective beta-cell numbers and/or function underlie essentially all major forms of diabetes and must be restored if diabetes is to be cured. Thus, the identification of the molecular regulators of beta-cell mass and a better understanding of the processes of beta-cell differentiation and proliferation may provide further insight for the development of new therapeutic targets for diabetes. This review will focus on the principal hormones and nutrients, as well as downstream signalling pathways regulating beta-cell mass in the adult. Furthermore, we will also address more recently appreciated regulators of beta-cell mass, such as microRNAs.
Collapse
Affiliation(s)
- Elena Tarabra
- School of Life Sciences, Warwick University, Gibbet Hill Road, Coventry CV4 7AL, UK
- *Elena Tarabra:
| | - Stella Pelengaris
- School of Life Sciences, Warwick University, Gibbet Hill Road, Coventry CV4 7AL, UK
| | - Michael Khan
- School of Life Sciences, Warwick University, Gibbet Hill Road, Coventry CV4 7AL, UK
| |
Collapse
|
155
|
Palau N, Rebuffat SA, Altirriba J, Piquer S, Hanzu FA, Gomis R, Barbera A. Role of IGFBP-3 in the regulation of β-cell mass during obesity: adipose tissue/β-cell cross talk. Endocrinology 2012; 153:177-87. [PMID: 22067319 DOI: 10.1210/en.2011-0181] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
In obesity an increase in β-cell mass occurs to cope with the rise in insulin demand. This β-cell plasticity is essential to avoid the onset of hyperglycemia, although the molecular mechanisms that regulate this process remain unclear. This study analyzed the role of adipose tissue in the control of β-cell replication. Using a diet-induced model of obesity, we obtained conditioned media from three different white adipose tissue depots. Only in the adipose tissue depot surrounding the pancreas did the diet induce changes that led to an increase in INS1E cells and the islet replication rate. To identify the factors responsible for this proliferative effect, adipose tissue gene expression analysis was conducted by microarrays and quantitative RT-PCR. Of all the differentially expressed proteins, only the secreted ones were studied. IGF binding protein 3 (Igfbp3) was identified as the candidate for this effect. Furthermore, in the conditioned media, although the blockage of IGFBP3 led to an increase in the proliferation rate, the blockage of IGF-I receptor decreased it. Taken together, these data show that obesity induces specific changes in the expression profile of the adipose tissue depot surrounding the pancreas, leading to a decrease in IGFBP3 secretion. This decrease acts in a paracrine manner, stimulating the β-cell proliferation rate, probably through an IGF-I-dependent mechanism. This cross talk between the visceral-pancreatic adipose tissue and β-cells is a novel mechanism that participates in the control of β-cell plasticity.
Collapse
Affiliation(s)
- Nuria Palau
- Diabetes and Obesity Laboratory, Institut d'Investigations Biomediques August Pi i Sunyer, Rosselló 149-153, E-08036 Barcelona, Spain
| | | | | | | | | | | | | |
Collapse
|
156
|
Lopez X, Cypess A, Manning R, O'Shea S, Kulkarni RN, Goldfine AB. Exogenous insulin enhances glucose-stimulated insulin response in healthy humans independent of changes in free fatty acids. J Clin Endocrinol Metab 2011; 96:3811-21. [PMID: 21956413 PMCID: PMC3232618 DOI: 10.1210/jc.2011-0627] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT Islet β-cells express both insulin receptors and insulin signaling proteins. Recent studies suggest insulin signaling is physiologically important for glucose sensing. OBJECTIVE Preexposure to insulin enhances glucose-stimulated insulin secretion (GSIS) in healthy humans. We evaluated whether the effect of insulin to potentiate GSIS is modulated through regulation of free fatty acids (FFA). DESIGN AND SETTING Subjects were studied on three occasions in this single-site study at an academic institution clinical research center. PATIENTS Subjects included nine healthy volunteers. INTERVENTIONS Glucose-induced insulin response was assessed on three occasions after 4 h saline (low insulin/sham) or isoglycemic-hyperinsulinemic (high insulin) clamps with or without intralipid and heparin infusion, using B28 Asp-insulin that could be distinguished from endogenous insulin immunologically. During the last 80 min of all three clamps, additional glucose was administered to stimulate insulin secretion (GSIS) with glucose concentrations maintained at similar concentrations during all studies. MAIN OUTCOME MEASURE β-Cell response to glucose stimulation was assessed. RESULTS Preexposure to exogenous insulin increased the endogenous insulin-secretory response to glucose by 32% compared with sham clamp (P = 0.001). This was accompanied by a drop in FFA during hyperinsulinemic clamp compared with the sham clamp (0.06 ± 0.02 vs. 0.60 ± 0.09 mEq/liter, respectively), which was prevented during the hyperinsulinemic clamp with intralipid/heparin infusion (1.27 ± 0.17 mEq/liter). After preexposure to insulin with intralipid/heparin infusion to maintain FFA concentration, GSIS was 21% higher compared with sham clamp (P < 0.04) and similar to preexposure to insulin without intralipid/heparin (P = 0.2). CONCLUSIONS Insulin potentiates glucose-stimulated insulin response independent of FFA concentrations in healthy humans.
Collapse
Affiliation(s)
- Ximena Lopez
- Joslin Diabetes Center and Harvard Medical School, Boston, Massachusetts 02215, USA
| | | | | | | | | | | |
Collapse
|
157
|
Wang YF, Khan M, van den Berg HA. Interaction of fast and slow dynamics in endocrine control systems with an application to β-cell dynamics. Math Biosci 2011; 235:8-18. [PMID: 22063267 DOI: 10.1016/j.mbs.2011.10.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2011] [Revised: 10/04/2011] [Accepted: 10/07/2011] [Indexed: 01/11/2023]
Abstract
Endocrine dynamics spans a wide range of time scales, from rapid responses to physiological challenges to with slow responses that adapt the system to the demands placed on it. We outline a non-linear averaging procedure to extract the slower dynamics in a way that accounts properly for the non-linear dynamics of the faster time scale and is applicable to a hierarchy of more than two time scales, although we restrict our discussion to two scales for the sake of clarity. The procedure is exact if the slow time scale is infinitely slow (the dimensionless ε-quantity is the period of the fast time scale fluctuation times an upper bound to the slow time scale rate of change). However, even for an imperfect separation of time scales we find that this construction provides an excellent approximation for the slow-time dynamics at considerably reduced computational cost. Besides the computation advantage, the averaged equation provided a qualitative insight into the interaction of the time scales. We demonstrate the procedure and its advantages by applying the theory to the model described by Tolić et al. [I.M. Tolić, E. Mosekilde, J. Sturis, Modeling the insulin-glucose feedback system: the significance of pulsatile insulin secretion, J. Theor. Biol. 207 (2000) 361-375.] for ultradian dynamics of the glucose-insulin homeostasis feedback system, extended to include β-cell dynamics. We find that the dynamics of the β-cell mass are dependent not only on the glycemic load (amount of glucose administered to the system), but also on the way this load is applied (i.e. three meals daily versus constant infusion), effects that are lost in the inappropriate methods used by the earlier authors. Furthermore, we find that the loss of the protection against apoptosis conferred by insulin that occurs at elevated levels of insulin has a functional role in keeping the β-cell mass in check without compromising regulatory function. We also find that replenishment of β-cells from a rapidly proliferating pool of cells, as opposed to the slow turn-over which characterises fully differentiated β-cells, is essential to the prevention of type 1 diabetes.
Collapse
Affiliation(s)
- Yi-Fang Wang
- Biological Sciences, University of Warwick, Coventry CV4 7AL, UK
| | | | | |
Collapse
|
158
|
Alejandro EU, Lim GE, Mehran AE, Hu X, Taghizadeh F, Pelipeychenko D, Baccarini M, Johnson JD. Pancreatic β-cell Raf-1 is required for glucose tolerance, insulin secretion, and insulin 2 transcription. FASEB J 2011; 25:3884-95. [PMID: 21817126 DOI: 10.1096/fj.10-180349] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Regulation of glucose homeostasis by insulin depends on pancreatic β-cell growth, survival, and function. Raf-1 kinase is a major downstream target of several growth factors that promote proliferation and survival of many cell types, including the pancreatic β cells. We have previously reported that insulin protects β cells from apoptosis and promotes proliferation by activating Raf-1 signaling in cultured human islets, mouse islets, and MIN6 cells. As Raf-1 activity is critical for basal apoptosis and insulin secretion in vitro, we hypothesized that Raf-1 may play an important role in glucose homeostasis in vivo. To test this hypothesis, we utilized the Cre-loxP recombination system to obtain a pancreatic β-cell-specific ablation of Raf-1 kinase gene (RIPCre(+/+):Raf-1(flox/flox)) and a complete set of littermate controls (RIPCre(+/+):Raf-1(wt/wt)). RIPCre(+/+):Raf-1(flox/flox) mice were viable, and no effects on weight gain were observed. RIPCre(+/+):Raf-1(flox/flox) mice had increased fasting blood glucose levels and impaired glucose tolerance but normal insulin tolerance compared to littermate controls. Insulin secretion in vivo and in isolated islets was markedly impaired, but there was no apparent effect on the exocytosis machinery. However, islet insulin protein and insulin 2 mRNA, but not insulin 1 mRNA, were dramatically reduced in Raf-1-knockout mice. Analysis of insulin 2 knockout mice demonstrated that this reduction in mRNA was sufficient to impair in vivo insulin secretion. Our data further indicate that Raf-1 specifically and acutely regulates insulin 2 mRNA via negative action on Foxo1, which has been shown to selectively control the insulin 2 gene. This work provides the first direct evidence that Raf-1 signaling is essential for the regulation of basal insulin transcription and the supply of releasable insulin in vivo.
Collapse
Affiliation(s)
- Emilyn U Alejandro
- Laboratory of Molecular Signalling in Diabetes, Diabetes Research Group, Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | | | | | | | | | | | | | | |
Collapse
|
159
|
Semple RK, Savage DB, Cochran EK, Gorden P, O'Rahilly S. Genetic syndromes of severe insulin resistance. Endocr Rev 2011; 32:498-514. [PMID: 21536711 DOI: 10.1210/er.2010-0020] [Citation(s) in RCA: 218] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Insulin resistance is among the most prevalent endocrine derangements in the world, and it is closely associated with major diseases of global reach including diabetes mellitus, atherosclerosis, nonalcoholic fatty liver disease, and ovulatory dysfunction. It is most commonly found in those with obesity but may also occur in an unusually severe form in rare patients with monogenic defects. Such patients may loosely be grouped into those with primary disorders of insulin signaling and those with defects in adipose tissue development or function (lipodystrophy). The severe insulin resistance of both subgroups puts patients at risk of accelerated complications and poses severe challenges in clinical management. However, the clinical disorders produced by different genetic defects are often biochemically and clinically distinct and are associated with distinct risks of complications. This means that optimal management of affected patients should take into account the specific natural history of each condition. In clinical practice, they are often underdiagnosed, however, with low rates of identification of the underlying genetic defect, a problem compounded by confusing and overlapping nomenclature and classification. We now review recent developments in understanding of genetic forms of severe insulin resistance and/or lipodystrophy and suggest a revised classification based on growing knowledge of the underlying pathophysiology.
Collapse
Affiliation(s)
- Robert K Semple
- Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom.
| | | | | | | | | |
Collapse
|
160
|
Abstract
Insulin resistance is the most important pathophysiological feature in many pre-diabetic states. Type 2 diabetes mellitus is a complex metabolic disease and its pathogenesis involves abnormalities in both peripheral insulin action and insulin secretion by pancreatic beta cells. The creation of monogenic or polygenic genetically manipulated mice models in a tissue-specific manner was of great help to elucidate the tissue-specificity of insulin action and its contribution to the overall insulin resistance. However, complete understanding of the molecular bases of the insulin action and resistance requires the identification of the intracellular pathways that regulate insulin-stimulated proliferation, differentiation and metabolism. Accordingly, cell lines derived from insulin target tissues such as brown adipose tissue, liver and beta islets lacking insulin receptors or sensitive candidate genes such as IRS-1, IRS-2, IRS-3, IR and PTP1B were developed. Indeed, these cell lines have been also very useful to understand the tissue-specificity of insulin action and inaction.
Collapse
Affiliation(s)
- Manuel Benito
- Departamento de Bioquímica y Biología Molecular II, Facultad de Farmacia, Universidad Complutense, Madrid, Spain.
| |
Collapse
|
161
|
Wu Y, Liu C, Sun H, Vijayakumar A, Giglou PR, Qiao R, Oppenheimer J, Yakar S, LeRoith D. Growth hormone receptor regulates β cell hyperplasia and glucose-stimulated insulin secretion in obese mice. J Clin Invest 2011; 121:2422-6. [PMID: 21555853 DOI: 10.1172/jci45027] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2010] [Accepted: 03/16/2011] [Indexed: 12/16/2022] Open
Abstract
Insulin, growth hormone (GH), and insulin-like growth factor-1 (IGF-1) play key roles in the regulation of β cell growth and function. Although β cells express the GH receptor, the direct effects of GH on β cells remain largely unknown. Here we have employed a rat insulin II promoter-driven (RIP-driven) Cre recombinase to disrupt the GH receptor in β cells (βGHRKO). βGHRKO mice fed a standard chow diet exhibited impaired glucose-stimulated insulin secretion but had no changes in β cell mass. When challenged with a high-fat diet, βGHRKO mice showed evidence of a β cell secretory defect, with further deterioration of glucose homeostasis indicated by their altered glucose tolerance and blunted glucose-stimulated insulin secretion. Interestingly, βGHRKO mice were impaired in β cell hyperplasia in response to a high-fat diet, with decreased β cell proliferation and overall reduced β cell mass. Therefore, GH receptor plays critical roles in glucose-stimulated insulin secretion and β cell compensation in response to a high-fat diet.
Collapse
Affiliation(s)
- Yingjie Wu
- Division of Endocrinology, Diabetes and Bone Disease, Department of Medicine, Mount Sinai School of Medicine, New York, New York 10029, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
162
|
Di Paola R, Caporarello N, Marucci A, Dimatteo C, Iadicicco C, Del Guerra S, Prudente S, Sudano D, Miele C, Parrino C, Piro S, Beguinot F, Marchetti P, Trischitta V, Frittitta L. ENPP1 affects insulin action and secretion: evidences from in vitro studies. PLoS One 2011; 6:e19462. [PMID: 21573217 PMCID: PMC3088669 DOI: 10.1371/journal.pone.0019462] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2011] [Accepted: 03/30/2011] [Indexed: 12/20/2022] Open
Abstract
The aim of this study was to deeper investigate the mechanisms through which
ENPP1, a negative modulator of insulin receptor (IR) activation, plays a role on
insulin signaling, insulin secretion and eventually glucose metabolism. ENPP1
cDNA (carrying either K121 or Q121 variant) was transfected in HepG2 liver-, L6
skeletal muscle- and INS1E beta-cells. Insulin-induced IR-autophosphorylation
(HepG2, L6, INS1E), Akt-Ser473,
ERK1/2-Thr202/Tyr204 and GSK3-beta Ser9
phosphorylation (HepG2, L6), PEPCK mRNA levels (HepG2) and
2-deoxy-D-glucose uptake (L6) was studied. GLUT 4 mRNA
(L6), insulin secretion and caspase-3 activation (INS1E) were also investigated.
Insulin-induced IR-autophosphorylation was decreased in HepG2-K, L6-K, INS1E-K
(20%, 52% and 11% reduction vs. untransfected cells) and
twice as much in HepG2-Q, L6-Q, INS1E-Q (44%, 92% and 30%).
Similar data were obtained with Akt-Ser473,
ERK1/2-Thr202/Tyr204 and GSK3-beta Ser9 in
HepG2 and L6. Insulin-induced reduction of PEPCK mRNA was progressively lower in
untransfected, HepG2-K and HepG2-Q cells (65%, 54%, 23%).
Insulin-induced glucose uptake in untransfected L6 (60% increase over
basal), was totally abolished in L6-K and L6-Q cells. GLUT 4 mRNA was slightly
reduced in L6-K and twice as much in L6-Q (13% and 25% reduction
vs. untransfected cells). Glucose-induced insulin secretion was 60%
reduced in INS1E-K and almost abolished in INS1E-Q. Serum deficiency activated
caspase-3 by two, three and four folds in untransfected INS1E, INS1E-K and
INS1E-Q. Glyburide-induced insulin secretion was reduced by 50% in
isolated human islets from homozygous QQ donors as compared to those from KK and
KQ individuals. Our data clearly indicate that ENPP1, especially when the Q121
variant is operating, affects insulin signaling and glucose metabolism in
skeletal muscle- and liver-cells and both function and survival of insulin
secreting beta-cells, thus representing a strong pathogenic factor predisposing
to insulin resistance, defective insulin secretion and glucose metabolism
abnormalities.
Collapse
Affiliation(s)
- Rosa Di Paola
- Research Unit of Diabetes and Endocrine
Diseases, IRCCS “Casa Sollievo della Sofferenza”, San Giovanni
Rotondo, Italy
- * E-mail: (RDP); (VT); (LF)
| | - Nunzia Caporarello
- Unit of Endocrinology, Department of Clinical
and Molecular Biomedicine, University of Catania Medical School, Garibaldi
Hospital, Catania, Italy
| | - Antonella Marucci
- Research Unit of Diabetes and Endocrine
Diseases, IRCCS “Casa Sollievo della Sofferenza”, San Giovanni
Rotondo, Italy
| | - Claudia Dimatteo
- Research Unit of Diabetes and Endocrine
Diseases, IRCCS “Casa Sollievo della Sofferenza”, San Giovanni
Rotondo, Italy
| | - Claudia Iadicicco
- Dipartimento di Biologia e Patologia Cellulare
e Molecolare and Istituto di Endocrinologia ed Oncologia Sperimentale del CNR,
Università degli Studi di Napoli Federico II, Naples, Italy
| | - Silvia Del Guerra
- Department of Endocrinology and Metabolism,
University of Pisa, Pisa, Italy
| | - Sabrina Prudente
- IRCCS “Casa Sollievo della Sofferenza,
Mendel Laboratory”, San Giovanni Rotondo, Italy
| | - Dora Sudano
- Unit of Endocrinology, Department of Clinical
and Molecular Biomedicine, University of Catania Medical School, Garibaldi
Hospital, Catania, Italy
| | - Claudia Miele
- Dipartimento di Biologia e Patologia Cellulare
e Molecolare and Istituto di Endocrinologia ed Oncologia Sperimentale del CNR,
Università degli Studi di Napoli Federico II, Naples, Italy
| | - Cristina Parrino
- Unit of Endocrinology, Department of Clinical
and Molecular Biomedicine, University of Catania Medical School, Garibaldi
Hospital, Catania, Italy
| | - Salvatore Piro
- Unit of Internal Medicine, Department of
Clinical and Molecular Biomedicine, University of Catania Medical School,
Garibaldi Hospital, Catania, Italy
| | - Francesco Beguinot
- Dipartimento di Biologia e Patologia Cellulare
e Molecolare and Istituto di Endocrinologia ed Oncologia Sperimentale del CNR,
Università degli Studi di Napoli Federico II, Naples, Italy
| | - Piero Marchetti
- Department of Endocrinology and Metabolism,
University of Pisa, Pisa, Italy
| | - Vincenzo Trischitta
- Research Unit of Diabetes and Endocrine
Diseases, IRCCS “Casa Sollievo della Sofferenza”, San Giovanni
Rotondo, Italy
- IRCCS “Casa Sollievo della Sofferenza,
Mendel Laboratory”, San Giovanni Rotondo, Italy
- Department of Experimental Medicine, Sapienza
University, Rome, Italy
- * E-mail: (RDP); (VT); (LF)
| | - Lucia Frittitta
- Unit of Endocrinology, Department of Clinical
and Molecular Biomedicine, University of Catania Medical School, Garibaldi
Hospital, Catania, Italy
- * E-mail: (RDP); (VT); (LF)
| |
Collapse
|
163
|
Porat S, Weinberg-Corem N, Tornovsky-Babaey S, Schyr-Ben-Haroush R, Hija A, Stolovich-Rain M, Dadon D, Granot Z, Ben-Hur V, White P, Girard CA, Karni R, Kaestner KH, Ashcroft FM, Magnuson MA, Saada A, Grimsby J, Glaser B, Dor Y. Control of pancreatic β cell regeneration by glucose metabolism. Cell Metab 2011; 13:440-449. [PMID: 21459328 DOI: 10.1016/j.cmet.2011.02.012] [Citation(s) in RCA: 247] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2010] [Revised: 01/12/2011] [Accepted: 02/23/2011] [Indexed: 01/09/2023]
Abstract
Recent studies revealed a surprising regenerative capacity of insulin-producing β cells in mice, suggesting that regenerative therapy for human diabetes could in principle be achieved. Physiologic β cell regeneration under stressed conditions relies on accelerated proliferation of surviving β cells, but the factors that trigger and control this response remain unclear. Using islet transplantation experiments, we show that β cell mass is controlled systemically rather than by local factors such as tissue damage. Chronic changes in β cell glucose metabolism, rather than blood glucose levels per se, are the main positive regulator of basal and compensatory β cell proliferation in vivo. Intracellularly, genetic and pharmacologic manipulations reveal that glucose induces β cell replication via metabolism by glucokinase, the first step of glycolysis, followed by closure of K(ATP) channels and membrane depolarization. Our data provide a molecular mechanism for homeostatic control of β cell mass by metabolic demand.
Collapse
Affiliation(s)
- Shay Porat
- Department of Developmental Biology and Cancer Research, The Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel; Department of Obstetrics and Gynecology, Hadassah-Hebrew University Medical Center, Mount Scopus, Jerusalem 91240, Israel
| | - Noa Weinberg-Corem
- Department of Developmental Biology and Cancer Research, The Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel
| | - Sharona Tornovsky-Babaey
- Endocrinology and Metabolism Service, Department of Internal Medicine, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel
| | - Rachel Schyr-Ben-Haroush
- Department of Developmental Biology and Cancer Research, The Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel; Endocrinology and Metabolism Service, Department of Internal Medicine, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel
| | - Ayat Hija
- Department of Developmental Biology and Cancer Research, The Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel
| | - Miri Stolovich-Rain
- Department of Developmental Biology and Cancer Research, The Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel
| | - Daniela Dadon
- Department of Developmental Biology and Cancer Research, The Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel
| | - Zvi Granot
- Department of Developmental Biology and Cancer Research, The Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel
| | - Vered Ben-Hur
- Department of Biochemistry and Molecular Biology, The Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel
| | - Peter White
- Department of Genetics and Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Christophe A Girard
- Department of Physiology, Anatomy, and Genetics, Oxford University, Oxford OX1 3QX, UK
| | - Rotem Karni
- Department of Biochemistry and Molecular Biology, The Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel
| | - Klaus H Kaestner
- Department of Genetics and Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Frances M Ashcroft
- Department of Physiology, Anatomy, and Genetics, Oxford University, Oxford OX1 3QX, UK
| | - Mark A Magnuson
- Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, TN 37232-0494, USA; Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232-0494, USA
| | - Ann Saada
- Department of Genetics and Metabolic Diseases, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel
| | - Joseph Grimsby
- Department of Metabolic Diseases, Hoffmann-La Roche, Nutley, NJ 07110, USA
| | - Benjamin Glaser
- Endocrinology and Metabolism Service, Department of Internal Medicine, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel.
| | - Yuval Dor
- Department of Developmental Biology and Cancer Research, The Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel.
| |
Collapse
|
164
|
Yang YHC, Szabat M, Bragagnini C, Kott K, Helgason CD, Hoffman BG, Johnson JD. Paracrine signalling loops in adult human and mouse pancreatic islets: netrins modulate beta cell apoptosis signalling via dependence receptors. Diabetologia 2011; 54:828-42. [PMID: 21212933 DOI: 10.1007/s00125-010-2012-5] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2010] [Accepted: 11/08/2010] [Indexed: 01/08/2023]
Abstract
AIMS/HYPOTHESIS Adult pancreatic islets contain multiple cell types that produce and secrete well characterised hormones, including insulin, glucagon and somatostatin. Although it is increasingly apparent that islets release and respond to more secreted factors than previously thought, systematic analyses are lacking. We therefore sought to identify potential autocrine and/or paracrine islet growth factor loops, and to characterise the function of the netrin family of islet-secreted factors and their receptors, which have been previously unreported in adult islets. METHODS Gene expression databases, islet-specific tag sequencing libraries and microarray datasets of FACS purified beta cells were used to compile a list of secreted factors and receptors present in mouse or human islets. Netrins and their receptors were further assessed using RT-PCR, Western blot analysis and immunofluorescence staining. The roles of netrin-1 and netrin-4 in beta cell function, apoptosis and proliferation were also examined. RESULTS We identified 233 secreted factors and 234 secreted factor receptors in islets. The presence of netrins and their receptors was further confirmed. Downregulation of caspase-3 activation was observed when MIN6 cells were exposed to exogenous netrin-1 and netrin-4 under hyperglycaemic conditions. Reduction in caspase-3 cleavage was linked to the decrease in dependence receptors, neogenin and unc-5 homologue A, as well as the activation of Akt and extracellular signal-regulated protein kinase (ERK) signalling. CONCLUSIONS/INTERPRETATION Our results highlight the large number of potential islet growth factors and point to a context-dependent pro-survival role for netrins in adult beta cells. Since diabetes results from a deficiency in functional beta cell mass, these studies are important steps towards developing novel therapies to improve beta cell survival.
Collapse
Affiliation(s)
- Y H C Yang
- Department of Cellular and Physiological Sciences, University of British Columbia, 5358 Life Sciences Building, 2350 Health Sciences Mall, Vancouver, BC, Canada
| | | | | | | | | | | | | |
Collapse
|
165
|
Hinault C, Kawamori D, Liew CW, Maier B, Hu J, Keller SR, Mirmira RG, Scrable H, Kulkarni RN. Δ40 Isoform of p53 controls β-cell proliferation and glucose homeostasis in mice. Diabetes 2011; 60:1210-22. [PMID: 21357466 PMCID: PMC3064094 DOI: 10.2337/db09-1379] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Investigating the dynamics of pancreatic β-cell mass is critical for developing strategies to treat both type 1 and type 2 diabetes. p53, a key regulator of the cell cycle and apoptosis, has mostly been a focus of investigation as a tumor suppressor. Although p53 alternative transcripts can modulate p53 activity, their functions are not fully understood. We hypothesized that β-cell proliferation and glucose homeostasis were controlled by Δ40p53, a p53 isoform lacking the transactivation domain of the full-length protein that modulates total p53 activity and regulates organ size and life span in mice. RESEARCH DESIGN AND METHODS We phenotyped metabolic parameters in Δ40p53 transgenic (p44tg) mice and used quantitative RT-PCR, Western blotting, and immunohistochemistry to examine β-cell proliferation. RESULTS Transgenic mice with an ectopic p53 gene encoding Δ40p53 developed hypoinsulinemia and glucose intolerance by 3 months of age, which worsened in older mice and led to overt diabetes and premature death from ∼14 months of age. Consistent with a dramatic decrease in β-cell mass and reduced β-cell proliferation, lower expression of cyclin D2 and pancreatic duodenal homeobox-1, two key regulators of proliferation, was observed, whereas expression of the cell cycle inhibitor p21, a p53 target gene, was increased. CONCLUSIONS These data indicate a significant and novel role for Δ40p53 in β-cell proliferation with implications for the development of age-dependent diabetes.
Collapse
Affiliation(s)
- Charlotte Hinault
- Research Division, Joslin Diabetes Center and Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Dan Kawamori
- Research Division, Joslin Diabetes Center and Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Chong Wee Liew
- Research Division, Joslin Diabetes Center and Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Bernhard Maier
- Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana
| | - Jiang Hu
- Research Division, Joslin Diabetes Center and Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | | | - Raghavendra G. Mirmira
- Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana
| | - Heidi Scrable
- Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota
| | - Rohit N. Kulkarni
- Research Division, Joslin Diabetes Center and Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
- Corresponding author: Rohit N. Kulkarni,
| |
Collapse
|
166
|
Abstract
Insulin resistance is the most important pathophysiological feature in many pre-diabetic states. Type 2 diabetes mellitus is a complex metabolic disease and its pathogenesis involves abnormalities in both peripheral insulin action and insulin secretion by pancreatic β-cells. The creation of monogenic or polygenic genetically manipulated mice models in a tissue-specific manner was of great help to elucidate the tissue specificity of insulin action and its contribution to the overall insulin resistance. However, a complete understanding of the molecular bases of insulin action and resistance requires the identification of intracellular pathways that regulate insulin-stimulated proliferation, differentiation and metabolism. Accordingly, cell lines derived from insulin target tissues such as brown adipose tissue, liver and beta islets lacking insulin resistance or sensitive candidate genes such as IRS-1, IRS-2, IRS-3, IR and PTP1B have been developed. Indeed, these cell lines have also been very useful to understand the tissue specificity of insulin action and inaction. Obesity is a risk factor for several components of the metabolic syndromes such as type 2 diabetes, dyslipidaemia and systolic hypertension, because white and brown adipose tissues as endocrine organs express and secrete a variety of adipocytokines that can act at both local and systemic levels, modulating the insulin sensitivity. Recent studies revealed that the subjects with the highest transcription rates of genes encoding TNF-α and IL-6 were prone to develop obesity, insulin resistance and type 2 diabetes. Accordingly, we specifically focus in this review on the impact of those adipocytokines on the modulation of insulin action in skeletal muscle.
Collapse
Affiliation(s)
- M Benito
- Departamento de Bioquímica y Biología Molecular II, Facultad de Farmacia, Universidad Complutense, Madrid, Spain.
| |
Collapse
|
167
|
Lin HV, Ren H, Samuel VT, Lee HY, Lu TY, Shulman GI, Accili D. Diabetes in mice with selective impairment of insulin action in Glut4-expressing tissues. Diabetes 2011; 60:700-9. [PMID: 21266328 PMCID: PMC3046830 DOI: 10.2337/db10-1056] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
OBJECTIVE Impaired insulin-dependent glucose disposal in muscle and fat is a harbinger of type 2 diabetes, but murine models of selective insulin resistance at these two sites are conspicuous by their failure to cause hyperglycemia. A defining feature of muscle and fat vis-à-vis insulin signaling is that they both express the insulin-sensitive glucose transporter Glut4. We hypothesized that diabetes is the result of impaired insulin signaling in all Glut4-expressing tissues. RESEARCH DESIGN AND METHODS To test the hypothesis, we generated mice lacking insulin receptors at these sites ("GIRKO" mice), including muscle, fat, and a subset of Glut4-positive neurons scattered throughout the central nervous system. RESULTS GIRKO mice develop diabetes with high frequency because of reduced glucose uptake in peripheral organs, excessive hepatic glucose production, and β-cell failure. CONCLUSIONS The conceptual advance of the present findings lies in the identification of a tissue constellation that melds cell-autonomous mechanisms of insulin resistance (in muscle/fat) with cell-nonautonomous mechanisms (in liver and β-cell) to cause overt diabetes. The data are consistent with the identification of Glut4 neurons as a distinct neuroanatomic entity with a likely metabolic role.
Collapse
Affiliation(s)
- Hua V. Lin
- Department of Medicine, Columbia University, New York, New York
| | - Hongxia Ren
- Department of Medicine, Columbia University, New York, New York
| | - Varman T. Samuel
- Department of Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Hui-Young Lee
- Department of Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Taylor Y. Lu
- Department of Medicine, Columbia University, New York, New York
| | - Gerald I. Shulman
- Department of Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Domenico Accili
- Department of Medicine, Columbia University, New York, New York
- Corresponding author: Domenico Accili,
| |
Collapse
|
168
|
Insulin and glucagon regulate pancreatic α-cell proliferation. PLoS One 2011; 6:e16096. [PMID: 21283589 PMCID: PMC3026810 DOI: 10.1371/journal.pone.0016096] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2010] [Accepted: 12/08/2010] [Indexed: 02/07/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) results from insulin resistance and β-cell dysfunction, in the setting of hyperglucagonemia. Glucagon is a 29 amino acid peptide hormone, which is secreted from pancreatic α cells: excessively high circulating levels of glucagon lead to excessive hepatic glucose output. We investigated if α-cell numbers increase in T2DM and what factor (s) regulate α-cell turnover. Leprdb/Leprdb (db/db) mice were used as a T2DM model and αTC1 cells were used to study potential α-cell trophic factors. Here, we demonstrate that in db/db mice α-cell number and plasma glucagon levels increased as diabetes progressed. Insulin treatment (EC50 = 2 nM) of α cells significantly increased α-cell proliferation in a concentration-dependent manner compared to non-insulin-treated α cells. Insulin up-regulated α-cell proliferation through the IR/IRS2/AKT/mTOR signaling pathway, and increased insulin-mediated proliferation was prevented by pretreatment with rapamycin, a specific mTOR inhibitor. GcgR antagonism resulted in reduced rates of cell proliferation in αTC1 cells. In addition, blockade of GcgRs in db/db mice improved glucose homeostasis, lessened α-cell proliferation, and increased intra-islet insulin content in β cells in db/db mice. These studies illustrate that pancreatic α-cell proliferation increases as diabetes develops, resulting in elevated plasma glucagon levels, and both insulin and glucagon are trophic factors to α-cells. Our current findings suggest that new therapeutic strategies for the treatment of T2DM may include targeting α cells and glucagon.
Collapse
|
169
|
Lin HV, Accili D. Reconstitution of insulin action in muscle, white adipose tissue, and brain of insulin receptor knock-out mice fails to rescue diabetes. J Biol Chem 2011; 286:9797-804. [PMID: 21239487 DOI: 10.1074/jbc.m110.210807] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Type 2 diabetes results from an impairment of insulin action. The first demonstrable abnormality of insulin signaling is a decrease of insulin-dependent glucose disposal followed by an increase in hepatic glucose production. In an attempt to dissect the relative importance of these two changes in disease progression, we have employed genetic knock-outs/knock-ins of the insulin receptor. Previously, we demonstrated that insulin receptor knock-out mice (Insr(-/-)) could be rescued from diabetes by reconstitution of insulin signaling in liver, brain, and pancreatic β cells (L1 mice). In this study, we used a similar approach to reconstitute insulin signaling in tissues that display insulin-dependent glucose uptake. Using GLUT4-Cre mice, we restored InsR expression in muscle, fat, and brain of Insr(-/-) mice (GIRKI (Glut4-insulin receptor knock-in line 1) mice). Unlike L1 mice, GIRKI mice failed to thrive and developed diabetes, although their survival was modestly extended when compared with Insr(-/-). The data underscore the role of developmental factors in the presentation of murine diabetes. The broader implication of our findings is that diabetes treatment should not necessarily target the same tissues that are responsible for disease pathogenesis.
Collapse
Affiliation(s)
- Hua V Lin
- Department of Medicine, Columbia University, New York, New York 10032, USA
| | | |
Collapse
|
170
|
Matschinsky FM, Zelent B, Doliba NM, Kaestner KH, Vanderkooi JM, Grimsby J, Berthel SJ, Sarabu R. Research and development of glucokinase activators for diabetes therapy: theoretical and practical aspects. Handb Exp Pharmacol 2011:357-401. [PMID: 21484579 DOI: 10.1007/978-3-642-17214-4_15] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Glucokinase Glucokinase (GK GK ; EC 2.7.1.1.) phosphorylates and regulates glucose metabolism in insulin-producing pancreatic beta-cells, hepatocytes, and certain cells of the endocrine and nervous systems allowing it to play a central role in glucose homeostasis glucose homeostasis . Most importantly, it serves as glucose sensor glucose sensor in pancreatic beta-cells mediating glucose-stimulated insulin biosynthesis and release and it governs the capacity of the liver to convert glucose to glycogen. Activating and inactivating mutations of the glucokinase gene cause autosomal dominant hyperinsulinemic hypoglycemia and hypoinsulinemic hyperglycemia in humans, respectively, illustrating the preeminent role of glucokinase in the regulation of blood glucose and also identifying the enzyme as a potential target for developing antidiabetic drugs antidiabetic drugs . Small molecules called glucokinase activators (GKAs) glucokinase activators (GKAs) which bind to an allosteric activator allosteric activator site of the enzyme have indeed been discovered and hold great promise as new antidiabetic agents. GKAs increase the enzyme's affinity for glucose and also its maximal catalytic rate. Consequently, they stimulate insulin biosynthesis and secretion, enhance hepatic glucose uptake, and augment glucose metabolism and related processes in other glucokinase-expressing cells. Manifestations of these effects, most prominently a lowering of blood glucose, are observed in normal laboratory animals and man but also in animal models of diabetes and patients with type 2 diabetes mellitus (T2DM T2DM ) type 2 diabetes mellitus (T2DM) . These compelling concepts and results sustain a strong R&D effort by many pharmaceutical companies to generate GKAs with characteristics allowing for a novel drug treatment of T2DM.
Collapse
Affiliation(s)
- Franz M Matschinsky
- Department of Biochemistry and Biophysics, University of Pennsylvania, Institute for Diabetes, Obesity and Metabolism, 415 Curie Blvd, 605 CRB, Philadelphia, PA 19104, USA.
| | | | | | | | | | | | | | | |
Collapse
|
171
|
Xuan S, Szabolcs M, Cinti F, Perincheri S, Accili D, Efstratiadis A. Genetic analysis of type-1 insulin-like growth factor receptor signaling through insulin receptor substrate-1 and -2 in pancreatic beta cells. J Biol Chem 2010; 285:41044-50. [PMID: 20947509 DOI: 10.1074/jbc.m110.144790] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Signaling by receptor tyrosine kinases regulates pancreatic β cell function. Inactivation of insulin receptor (InsR), IGF1 receptor (Igf1r), or Irs1 in β cells impairs insulin secretion. Conversely, Irs2 ablation impairs β cell replication. In this study, we examined aspects of the Igf1r regulatory signaling cascade in β cells. To examine genetically the involvement of Irs1 and Irs2 in Igf1r signaling, we generated double mutant mice lacking Igf1r specifically in pancreatic β cells in an Irs1- or Irs2-null background. We show that Igf1r/Irs1 double mutants do not differ phenotypically from Irs1 single mutants and exhibit hyperinsulinemia, while maintaining normal β cell mass and glucose tolerance. In contrast, lack of Igf1r function in β cells aggravates the consequences of Irs2 ablation in double mutants and results in lethal diabetes by 6 weeks of age. This additivity of phenotypic manifestations indicates that Irs2 serves a pathway that is largely independent of Igf1r signaling. Consistent with the view that the latter is the InsR pathway, we show that combined β cell-specific knock-out of both Insr and Igf1r results in a phenocopy of double mutants lacking Igf1r and Irs2. We conclude that Igf1r signals primarily through Irs1 and affects insulin secretion, whereas β cell proliferation is mainly regulated by InsR using Irs2 as a downstream signaling effector. The insulin and IGF pathways appear to control β cell functions independently and selectively.
Collapse
Affiliation(s)
- Shouhong Xuan
- Department of Genetics and Development, College of Physicians & Surgeons of Columbia University, New York, New York 10032, USA
| | | | | | | | | | | |
Collapse
|
172
|
Lebrun P, Cognard E, Gontard P, Bellon-Paul R, Filloux C, Berthault MF, Magnan C, Ruberte J, Luppo M, Pujol A, Pachera N, Herchuelz A, Bosch F, Van Obberghen E. The suppressor of cytokine signalling 2 (SOCS2) is a key repressor of insulin secretion. Diabetologia 2010; 53:1935-46. [PMID: 20499047 DOI: 10.1007/s00125-010-1786-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2010] [Accepted: 04/01/2010] [Indexed: 10/19/2022]
Abstract
AIMS/HYPOTHESIS Suppressor of cytokine signalling (SOCS) proteins are powerful inhibitors of pathways involved in survival and function of pancreatic beta cells. Whereas SOCS1 and SOCS3 have been involved in immune and inflammatory processes, respectively, in beta cells, nothing is known about SOCS2 implication in the pancreas. METHODS Transgenic (tg) mice were generated that constitutively produced SOCS2 in beta cells (betaSOCS2) to define whether this protein is implicated in beta cell functioning and/or survival. RESULTS Constitutive production of SOCS2 in beta cells leads to hyperglycaemia and glucose intolerance. This phenotype is not a consequence of decreased beta cell mass or inhibition of insulin synthesis. However, insulin secretion to various secretagogues is profoundly altered in intact animals and isolated islets. Interestingly, constitutive SOCS2 production dampens the rise in cytosolic free calcium concentration induced by glucose, while glucose metabolism is unchanged. Moreover, tg islets have a depletion in endoplasmic reticulum Ca(2+) stores, suggesting that SOCS2 interferes with calcium fluxes. Finally, in betaSOCS2 mice proinsulin maturation is impaired, leading to an altered structure of insulin secretory granules and augmented levels of proinsulin. The latter is likely to be due to decreased production of prohormone convertase 1 (PC1/3), which plays a key role in proinsulin cleavage. CONCLUSIONS/INTERPRETATIONS SOCS2 was shown to be a potent regulator of proinsulin processing and insulin secretion in beta cells. While its constitutive production is insufficient to induce overt diabetes in this mouse model, it causes glucose intolerance. Thus, increased SOCS2 production could be an important event predisposing to beta cell failure.
Collapse
Affiliation(s)
- P Lebrun
- INSERM U907, Avenue de Valombrose, Nice, France.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
173
|
Grouwels G, Cai Y, Hoebeke I, Leuckx G, Heremans Y, Ziebold U, Stangé G, Chintinne M, Ling Z, Pipeleers D, Heimberg H, Van de Casteele M. Ectopic expression of E2F1 stimulates beta-cell proliferation and function. Diabetes 2010; 59:1435-44. [PMID: 20299467 PMCID: PMC2874704 DOI: 10.2337/db09-1295] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
OBJECTIVE Generating functional beta-cells by inducing their proliferation may provide new perspectives for cell therapy in diabetes. Transcription factor E2F1 controls G(1)- to S-phase transition during the cycling of many cell types and is required for pancreatic beta-cell growth and function. However, the consequences of overexpression of E2F1 in beta-cells are unknown. RESEARCH DESIGN AND METHODS The effects of E2F1 overexpression on beta-cell proliferation and function were analyzed in isolated rat beta-cells and in transgenic mice. RESULTS Adenovirus AdE2F1-mediated overexpression of E2F1 increased the proliferation of isolated primary rat beta-cells 20-fold but also enhanced beta-cell death. Coinfection with adenovirus AdAkt expressing a constitutively active form of Akt (protein kinase B) suppressed beta-cell death to control levels. At 48 h after infection, the total beta-cell number and insulin content were, respectively, 46 and 79% higher in AdE2F1+AdAkt-infected cultures compared with untreated. Conditional overexpression of E2F1 in mice resulted in a twofold increase of beta-cell proliferation and a 70% increase of pancreatic insulin content, but did not increase beta-cell mass. Glucose-challenged insulin release was increased, and the mice showed protection against toxin-induced diabetes. CONCLUSIONS Overexpression of E2F1, either in vitro or in vivo, can stimulate beta-cell proliferation activity. In vivo E2F1 expression significantly increases the insulin content and function of adult beta-cells, making it a strategic target for therapeutic manipulation of beta-cell function.
Collapse
Affiliation(s)
- Gael Grouwels
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium; and
| | - Ying Cai
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium; and
| | - Inge Hoebeke
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium; and
| | - Gunter Leuckx
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium; and
| | - Yves Heremans
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium; and
| | - Ulrike Ziebold
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Geert Stangé
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium; and
| | - Marie Chintinne
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium; and
| | - Zhidong Ling
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium; and
| | - Daniel Pipeleers
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium; and
| | - Harry Heimberg
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium; and
- Corresponding author: Harry Heimberg,
| | | |
Collapse
|
174
|
Georgia S, Hinault C, Kawamori D, Hu J, Meyer J, Kanji M, Bhushan A, Kulkarni RN. Cyclin D2 is essential for the compensatory beta-cell hyperplastic response to insulin resistance in rodents. Diabetes 2010; 59:987-96. [PMID: 20103709 PMCID: PMC2844846 DOI: 10.2337/db09-0838] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
OBJECTIVE A major determinant of the progression from insulin resistance to the development of overt type 2 diabetes is a failure to mount an appropriate compensatory beta-cell hyperplastic response to maintain normoglycemia. We undertook the present study to directly explore the significance of the cell cycle protein cyclin D2 in the expansion of beta-cell mass in two different models of insulin resistance. RESEARCH DESIGN AND METHODS We created compound knockouts by crossing mice deficient in cyclin D2 (D2KO) with either the insulin receptor substrate 1 knockout (IRS1KO) mice or the insulin receptor liver-specific knockout mice (LIRKO), neither of which develops overt diabetes on its own because of robust compensatory beta-cell hyperplasia. We phenotyped the double knockouts and used RT-qPCR and immunohistochemistry to examine beta-cell mass. RESULTS Both compound knockouts, D2KO/LIRKO and D2KO/IRS1KO, exhibited insulin resistance and hyperinsulinemia and an absence of compensatory beta-cell hyperplasia. However, the diabetic D2KO/LIRKO group rapidly succumbed early compared with a relatively normal lifespan in the glucose-intolerant D2KO/IRS1KO mice. CONCLUSIONS This study provides direct genetic evidence that cyclin D2 is essential for the expansion of beta-cell mass in response to a spectrum of insulin resistance and points to the cell-cycle protein as a potential therapeutic target that can be harnessed for preventing and curing type 2 diabetes.
Collapse
Affiliation(s)
- Senta Georgia
- Larry Hillblom Islet Research Center, University of California, Los Angeles, David Geffen School of Medicine, Los Angeles, California
| | - Charlotte Hinault
- Joslin Diabetes Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Dan Kawamori
- Joslin Diabetes Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Jiang Hu
- Joslin Diabetes Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - John Meyer
- Joslin Diabetes Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Murtaza Kanji
- Larry Hillblom Islet Research Center, University of California, Los Angeles, David Geffen School of Medicine, Los Angeles, California
| | - Anil Bhushan
- Larry Hillblom Islet Research Center, University of California, Los Angeles, David Geffen School of Medicine, Los Angeles, California
- Corresponding author: Rohit N. Kulkarni, , or Anil Bhushan,
| | - Rohit N. Kulkarni
- Joslin Diabetes Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Corresponding author: Rohit N. Kulkarni, , or Anil Bhushan,
| |
Collapse
|
175
|
Fajas L, Blanchet E, Annicotte JS. CDK4, pRB and E2F1: connected to insulin. Cell Div 2010; 5:6. [PMID: 20181095 PMCID: PMC2829545 DOI: 10.1186/1747-1028-5-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2010] [Accepted: 02/05/2010] [Indexed: 01/09/2023] Open
Abstract
Pancreatic beta-cells are metabolic sensors involved in the control of glucose homeostasis. This particular cell type controls insulin secretion through a fine-tuned process, which dregulation have important pathological consequences, such as observed during type 2 diabetes. We recently implicated E2F1 in the control of glucose homeostasis. First we showed that E2f1-/- mice have decreased pancreatic size, as the result of impaired postnatal pancreatic growth. We observed in this study that E2F1 was highly expressed in non-proliferating pancreatic beta-cells, suggesting that E2F1, besides the control of beta-cell number could have a role in pancreatic beta-cell function. We demonstrate in our recent study, both in vitro and in vivo that E2F1 directly regulates the expression of Kir6.2, a key component of the KATP channel involved in the regulation of glucose-induced insulin secretion in pancreatic beta-cells. Expression of Kir6.2 is lost in pancreas of E2f1-/- mice, resulting in insulin secretion defects in these mice. Furthermore, we demonstrated by in tissue chromatin immunoprecipitation analysis that regulation of Kir6.2 expression by E2F1 follows the same regulatory pathway that the classical E2F1 target genes, implicating the participation of CDK4 and retinoblastoma protein. Moreover, in this context, E2F1 transcriptional activity is regulated by glucose and insulin through the CDK4-dependent inactivation of the pRB protein. In summary we provide evidence that the CDK4-pRB-E2F1 regulatory pathway is involved in glucose homeostasis. In our recent study we decipher a new function for these factors in the control of insulin secretion and open up new avenues for the treatment of metabolic diseases, in particular type 2 diabetes.
Collapse
Affiliation(s)
- Lluis Fajas
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, F-34298, France.
| | | | | |
Collapse
|
176
|
Robertson K, Dong J, De Jesus K, Liu JL. IGF-I overexpression does not promote compensatory islet cell growth in diet-induced obesity. Endocrine 2010; 37:47-54. [PMID: 19876774 DOI: 10.1007/s12020-009-9259-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2009] [Accepted: 09/11/2009] [Indexed: 01/20/2023]
Abstract
Although IGF-I was known to stimulate the growth of pancreatic islet cells from early in vitro experiments and in vivo reports on rodents, recent gene targeting experiments have indicated that IGF-I and its receptor do not play a major role in normal islet cell growth. In our previous reports, liver- or pancreatic-specific IGF-I deficiency caused no decrease in β-cell mass; a general and β-cell-enriched IGF-I overexpression caused no change in normal islet cell growth. On the other hand, increased metabolic demands (such as in obesity and insulin resistance) result in β-cell compensation in cell number and insulin secretion. In order to test whether IGF-I could promote islet cell growth and facilitate islet compensation due to obesity-induced insulin resistance, we have challenged MT-IGF mice to a high-fat diet. After 28 weeks, both MT-IGF mice and wild-type littermates gained comparable 40-57% of body weight, with similar increases in fat masses; all mice maintained a normal sensitivity to insulin and did not become severely hyperglycemic. Nevertheless, compared to wild-type littermates, the equally obese MT-IGF mice maintained improved glucose tolerance and a diminished insulin level; similar to when fed a normal chow diet. More importantly, under IGF-I overexpression, there was no further increase in β-cell mass caused by obesity. Thus, IGF-I overexpression had no significant effect on weight gain and islet cell compensation in response to high-fat diet-induced obesity.
Collapse
Affiliation(s)
- Katie Robertson
- Fraser Laboratories for Diabetes Research, Department of Medicine, McGill University Health Centre, Room H5-21, Royal Victoria Hospital, 687 Pine Avenue West, Montreal, QC H3A 1A1, Canada
| | | | | | | |
Collapse
|
177
|
Alejandro EU, Kalynyak TB, Taghizadeh F, Gwiazda KS, Rawstron EK, Jacob KJ, Johnson JD. Acute insulin signaling in pancreatic beta-cells is mediated by multiple Raf-1 dependent pathways. Endocrinology 2010; 151:502-12. [PMID: 20056832 PMCID: PMC2817610 DOI: 10.1210/en.2009-0678] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Insulin enhances the proliferation and survival of pancreatic beta-cells, but its mechanisms remain unclear. We hypothesized that Raf-1, a kinase upstream of both ERK and Bad, might be a critical target of insulin in beta-cells. To test this hypothesis, we treated human and mouse islets as well as MIN6 beta-cells with multiple insulin concentrations and examined putative downstream targets using immunoblotting, immunoprecipitation, quantitative fluorescent imaging, and cell death assays. Low doses of insulin rapidly activated Raf-1 by dephosphorylating serine 259 and phosphorylating serine 338 in human islets, mouse islets, and MIN6 cells. The phosphorylation of ERK by insulin was eliminated by exposure to a Raf inhibitor (GW5074) or transfection with a dominant-negative Raf-1 mutant. Insulin also enhanced the interaction between mitochondrial Raf-1 and Bcl-2 agonist of cell death (Bad), promoting Bad inactivation via its phosphorylation on serine 112. Insulin-stimulated ERK phosphorylation was abrogated by calcium chelation, calcineurin and calmodulin-dependent protein kinase II inhibitors, and Ned-19, a nicotinic acid adenine dinucleotide phosphate receptor (NAADPR) antagonist. Blocking Raf-1 and Ca(2+) signaling resulted in nonadditive beta-cell death. Autocrine insulin signaling partly accounted for the effects of glucose on ERK phosphorylation. Our results demonstrate that Raf-1 is a critical target of insulin in primary beta-cells. Activation of Raf-1 leads to both an ERK-dependent pathway that involves nicotinic acid adenine dinucleotide phosphate-sensitive Ca(2+) stores and Ca(2+)-dependent phosphorylation events, and an ERK-independent pathway that involves Bad inactivation at the mitochondria. Together our findings identify a novel insulin signaling pathway in beta-cells and shed light on insulin's antiapoptotic and mitogenic mechanisms.
Collapse
Affiliation(s)
- Emilyn U Alejandro
- Department of Cellular and Physiological Sciences, University of British Columbia, 5358 Life Sciences Building, 2350 Health Sciences Mall, Vancouver, British Columbia, Canada
| | | | | | | | | | | | | |
Collapse
|
178
|
Kolb H, Mandrup-Poulsen T. The global diabetes epidemic as a consequence of lifestyle-induced low-grade inflammation. Diabetologia 2010; 53:10-20. [PMID: 19890624 DOI: 10.1007/s00125-009-1573-7] [Citation(s) in RCA: 195] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2009] [Accepted: 09/24/2009] [Indexed: 02/08/2023]
Abstract
The recent major increase in the global incidence of type 2 diabetes suggests that most cases of this disease are caused by changes in environment and lifestyle. All major risk factors for type 2 diabetes (overnutrition, low dietary fibre, sedentary lifestyle, sleep deprivation and depression) have been found to induce local or systemic low-grade inflammation that is usually transient or milder in individuals not at risk for type 2 diabetes. By contrast, inflammatory responses to lifestyle factors are more pronounced and prolonged in individuals at risk of type 2 diabetes and appear to occur also in the pancreatic islets. Chronic low-grade inflammation will eventually lead to overt diabetes if counter-regulatory circuits to inflammation and metabolic stress are compromised because of a genetic and/or epigenetic predisposition. Hence, it is not the lifestyle change per se but a deficient counter-regulatory response in predisposed individuals which is crucial to disease pathogenesis. Novel approaches of intervention may target these deficient defence mechanisms.
Collapse
Affiliation(s)
- H Kolb
- Hagedorn Research Institute, DK 2820 Gentofte, Denmark.
| | | |
Collapse
|
179
|
Mechanisms of pancreatic beta-cell apoptosis in diabetes and its therapies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 654:447-62. [PMID: 20217509 DOI: 10.1007/978-90-481-3271-3_19] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Diabetes occurs when beta-cells no longer function properly or have been destroyed. Pancreatic beta-cell death by apoptosis contributes significantly in both autoimmune type 1 diabetes and type 2 diabetes. Pancreatic beta-cell death can be induced by multiple stresses in both major types of diabetes. There are also several rare forms of diabetes, including Wolcott-Rallison syndrome, Wolfram syndrome, as well as some forms of maturity onset diabetes of the young that are caused by mutations in genes that may play important roles in beta-cell survival. The use of islet transplantation as a treatment for diabetes is also limited by excessive beta-cell apoptosis. Mechanistic insights into the control of pancreatic beta-cell apoptosis are therefore important for the prevention and treatment of diabetes. Indeed, a substantial quantity of research has been dedicated to this area over the past decade. In this chapter, we review the factors that influence the propensity of beta-cells to undergo apoptosis and the mechanisms of this programmed cell death in the initiation and progression of diabetes.
Collapse
|
180
|
Golson M, Misfeldt AA, Kopsombut U, Petersen C, Gannon M. High Fat Diet Regulation of β-Cell Proliferation and β-Cell Mass. THE OPEN ENDOCRINOLOGY JOURNAL 2010; 4:10.2174/1874216501004010066. [PMID: 24339840 PMCID: PMC3856766 DOI: 10.2174/1874216501004010066] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Type 2 Diabetes (T2D) is characterized by relative insulin insufficiency, caused when peripheral tissues such as liver, muscle, and adipocytes have a decreased response to insulin. One factor that elevates the risk for insulin resistance and T2D is obesity. In obese patients without T2D and initially in people who develop T2D, pancreatic β-cells are able to compensate for insulin resistance by increasing β-cell mass, effected by increased proliferation and hypertrophy, as well as increased insulin secretion per β-cell. In patients that go on to develop T2D, however, this initial period of compensation is followed by β-cell failure due to decreased proliferation and increased apoptosis. The forkhead box transcription factor FoxM1 is required for β-cell replication in mice after four weeks of age, during pregnancy, and after partial pancreatectomy. We investigated whether it is also required for β-cell proliferation due to diet-induced obesity.
Collapse
Affiliation(s)
- M.L. Golson
- Department of Medicine, Division of Diabetes, Endocrinology, and Metabolism, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - A. Ackermann Misfeldt
- Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - U.G. Kopsombut
- Department of Medicine, Division of Diabetes, Endocrinology, and Metabolism, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - C.P. Petersen
- Department of Medicine, Division of Diabetes, Endocrinology, and Metabolism, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - M. Gannon
- Department of Medicine, Division of Diabetes, Endocrinology, and Metabolism, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
181
|
Morpurgo G, Fioretti B, Catacuzzeno L. The main product of specialized tissues regulates cell life and may cause neoplastic transformation. Med Hypotheses 2009; 74:847-54. [PMID: 20036074 DOI: 10.1016/j.mehy.2009.11.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2009] [Accepted: 11/24/2009] [Indexed: 11/15/2022]
Abstract
Many tissues and cells in vertebrates are highly specialized and devoted to a single function through the action of a single molecule, that we call the "main product" (MP) of the cell. The hypothesis here proposed is that these MPs control all aspects of the cell life, namely activity, division, differentiation and apoptosis. Evidences supporting this hypothesis are reported for the immune system, pancreatic beta-cells, melanocytes, connective tissues, thyroid cells, skin and erythroid cells. In all cases cell division and differentiation is promoted by a normal activity of the MP, while hyperactivity leads to cell apoptosis. Evidences are also provided that alterations of the activity of the MP may elicit pathological disorders; in particular mutations altering the structure of the MP may elicit tumoural transformation.
Collapse
Affiliation(s)
- Giorgio Morpurgo
- Dipartimento di Biologia Cellulare e Ambientale, Universita' di Perugia, Perugia, Italy
| | | | | |
Collapse
|
182
|
Meng Z, Lv J, Luo Y, Lin Y, Zhu Y, Nie J, Yang T, Sun Y, Han X. Forkhead box O1/pancreatic and duodenal homeobox 1 intracellular translocation is regulated by c-Jun N-terminal kinase and involved in prostaglandin E2-induced pancreatic beta-cell dysfunction. Endocrinology 2009; 150:5284-93. [PMID: 19837872 DOI: 10.1210/en.2009-0671] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Prostaglandin E(2) (PGE(2)) is a well-known mediator of beta-cell dysfunction in both type 1 and type 2 diabetes. We recently reported that down-regulation of the Akt pathway activity is implicated in PGE(2)-induced pancreatic beta-cell dysfunction. The aim of this study was to further dissect the signaling pathway of this process in pancreatic beta-cell line HIT-T15 cells and primary mouse islets. We found that PGE(2) time-dependently increased the c-Jun N-terminal kinase (JNK) pathway activity. JNK inhibition by the JNK-specific inhibitor SP600125 reversed PGE(2)-inhibited glucose-stimulated insulin secretion (GSIS). PGE(2) induced dephosphorylation of Akt and FOXO1, leading to nuclear localization and transactivation of FOXO1. Activation of FOXO1 induced nuclear exclusion but had no obvious effect on the whole-cell protein level of pancreatic and duodenal homeobox 1 (PDX1). However, these effects were all attenuated by JNK inhibition. Furthermore, adenovirus-mediated overexpression of dominant-negative (DN)-FOXO1 abolished whereas constitutively active (CA)-FOXO1 mimicked the effects of PGE(2) on GSIS in isolated mouse islets. In addition, we demonstrated that DN-JNK1 but not DN-JNK2 or CA-Akt abolished the PGE(2)-induced AP-1 luciferase reporter activity, whereas DN-JNK1 and CA-Akt but not DN-JNK2 reversed the effect of PGE(2) on FOXO1 transcriptional activity, and overexpression of DN-JNK1 rescued PGE(2)-impaired GSIS in mouse islets. Our results revealed that activation of the JNK is involved in PGE(2)-induced beta-cell dysfunction. PGE(2)-mediated JNK1 activation, through dephosphorylation of Akt and FOXO1, leads to nuclear accumulation of FOXO1 and nucleocytoplasmic shuttling of PDX1, finally resulting in defective GSIS in pancreatic beta-cells.
Collapse
Affiliation(s)
- Zhuoxian Meng
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Jiangsu Diabetes Centre, Nanjing Medical University, Nanjing 210029, China
| | | | | | | | | | | | | | | | | |
Collapse
|
183
|
Liu S, Okada T, Assmann A, Soto J, Liew CW, Bugger H, Shirihai OS, Abel ED, Kulkarni RN. Insulin signaling regulates mitochondrial function in pancreatic beta-cells. PLoS One 2009; 4:e7983. [PMID: 19956695 PMCID: PMC2776992 DOI: 10.1371/journal.pone.0007983] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2009] [Accepted: 10/26/2009] [Indexed: 12/18/2022] Open
Abstract
Insulin/IGF-I signaling regulates the metabolism of most mammalian tissues including pancreatic islets. To dissect the mechanisms linking insulin signaling with mitochondrial function, we first identified a mitochondria-tethering complex in β-cells that included glucokinase (GK), and the pro-apoptotic protein, BADS. Mitochondria isolated from β-cells derived from β-cell specific insulin receptor knockout (βIRKO) mice exhibited reduced BADS, GK and protein kinase A in the complex, and attenuated function. Similar alterations were evident in islets from patients with type 2 diabetes. Decreased mitochondrial GK activity in βIRKOs could be explained, in part, by reduced expression and altered phosphorylation of BADS. The elevated phosphorylation of p70S6K and JNK1 was likely due to compensatory increase in IGF-1 receptor expression. Re-expression of insulin receptors in βIRKO cells partially restored the stoichiometry of the complex and mitochondrial function. These data indicate that insulin signaling regulates mitochondrial function and have implications for β-cell dysfunction in type 2 diabetes.
Collapse
Affiliation(s)
- Siming Liu
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
- Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Terumasa Okada
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
- Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Anke Assmann
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
- Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Jamie Soto
- Division of Endocrinology, Metabolism and Diabetes and Program in Molecular Medicine, University of Utah School of Medicine, Salt Lake City, Utah, United States of America
| | - Chong Wee Liew
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
- Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Heiko Bugger
- Division of Endocrinology, Metabolism and Diabetes and Program in Molecular Medicine, University of Utah School of Medicine, Salt Lake City, Utah, United States of America
| | - Orian S. Shirihai
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - E. Dale Abel
- Division of Endocrinology, Metabolism and Diabetes and Program in Molecular Medicine, University of Utah School of Medicine, Salt Lake City, Utah, United States of America
| | - Rohit N. Kulkarni
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
- Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
184
|
Bonnesen C, Nelander GM, Hansen BF, Jensen P, Krabbe JS, Jensen MB, Hegelund AC, Svendsen JE, Oleksiewicz MB. Synchronization in G0/G1 enhances the mitogenic response of cells overexpressing the human insulin receptor A isoform to insulin. Cell Biol Toxicol 2009; 26:293-307. [PMID: 19898946 PMCID: PMC2896650 DOI: 10.1007/s10565-009-9142-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2009] [Accepted: 10/22/2009] [Indexed: 01/01/2023]
Abstract
Evaluating mitogenic signaling specifically through the human insulin receptor (IR) is relevant for the preclinical safety assessment of developmental insulin analogs. It is known that overexpression of IR sensitizes cells to the mitogenic effects of insulin, but it is essentially unknown how mitogenic responses can be optimized to allow practical use of such recombinant cell lines for preclinical safety testing. We constitutively overexpressed the short isoform of the human insulin receptor (hIR-A, exon 11-negative) in L6 rat skeletal myoblasts. Because the mitogenic effect of growth factors such as insulin is expected to act in G0/G1, promoting S-phase entry, we developed a combined topoinhibition + serum deprivation strategy to explore the effect of G0/G1 synchronization as an independent parameter in the context of serum deprivation, the latter being routinely used to reduce background in mitogenicity assays. G0/G1 synchronization significantly improved the mitogenic responses of L6-hIR cells to insulin, measured by 3H-thymidine incorporation. Comparison with the parental L6 cells using phospho-mitogen-activated protein kinase, phospho-AKT, as well as 3H-thymidine incorporation end points supported that the majority of the mitogenic effect of insulin in L6-hIR cells was mediated by the overexpressed hIR-A. Using the optimized L6-hIR assay, we found that the X-10 insulin analog was more mitogenic than native human insulin, supporting that X-10 exhibits increased mitogenic signaling through the hIR-A. In summary, this study provides the first demonstration that serum deprivation may not be sufficient, and G0/G1 synchronization may be required to obtain optimal responsiveness of hIR-overexpressing cell lines for preclinical safety testing.
Collapse
|
185
|
Abstract
Telomerase is a specialized reverse transcriptase that is responsible for extending and preserving the end of the chromosomes (telomeres). Telomerase plays a key role in regulating the lifespan of mammalian cells and is involved in critical aspects of cellular ageing processes. In this review, we will briefly summarize our current understanding of the functions of telomeres, telomerase and their regulation. Considering that compensatory islet hyperplasia and beta-cell regeneration play important roles in the prevention and/or delay of the onset of overt diabetes, we will also examine current literature regarding the effects of diabetes on telomere shortening and provide insights from our own studies on the role of telomerase in beta-cell regeneration.
Collapse
Affiliation(s)
- C W Liew
- Joslin Diabetes Center and Department of Medicine, Harvard Medical School, Boston, MA, USA
| | | | | |
Collapse
|
186
|
Talchai C, Lin HV, Kitamura T, Accili D. Genetic and biochemical pathways of beta-cell failure in type 2 diabetes. Diabetes Obes Metab 2009; 11 Suppl 4:38-45. [PMID: 19817787 DOI: 10.1111/j.1463-1326.2009.01115.x] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
We review mechanisms of beta-cell failure in type 2 diabetes. A wealth of information indicates that it is caused by impaired insulin secretion and decreased beta-cell mass. Interestingly, there appears to be a link between these two mechanisms. The earliest reaction to peripheral insulin resistance is an increase in insulin production, owing primarily to increased secretion, and to a lesser extent to decreased clearance. Experimental animal models indicate that hyperinsulinaemia promotes an increase in beta-cell mass, largely via increased beta-cell replication. In contrast, following the onset of overt diabetes, there is a slowly progressive loss of beta-cell function and mass, both in animal models and in diabetic humans. It is of great interest that most diabetes-associated genes identified in genome-wide association studies appear to be enriched in the beta-cell and to have the potential to regulate mass and/or function. Here, we review evidence derived from experimental animal models to unravel the mechanisms underlying beta-cell dysfunction. We focus primarily on signalling pathways, as opposed to nutrient sensing, and specifically on the notion that insulin and growth factor signalling via Foxo1 in pancreatic beta-cells links insulin secretion with cellular proliferation and survival.
Collapse
Affiliation(s)
- C Talchai
- Naomi Berrie Diabetes Center, Department of Medicine, College of Physicians and Surgeons of Columbia University, New York, NY, USA
| | | | | | | |
Collapse
|
187
|
Belfiore A, Frasca F, Pandini G, Sciacca L, Vigneri R. Insulin receptor isoforms and insulin receptor/insulin-like growth factor receptor hybrids in physiology and disease. Endocr Rev 2009; 30:586-623. [PMID: 19752219 DOI: 10.1210/er.2008-0047] [Citation(s) in RCA: 739] [Impact Index Per Article: 46.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
In mammals, the insulin receptor (IR) gene has acquired an additional exon, exon 11. This exon may be skipped in a developmental and tissue-specific manner. The IR, therefore, occurs in two isoforms (exon 11 minus IR-A and exon 11 plus IR-B). The most relevant functional difference between these two isoforms is the high affinity of IR-A for IGF-II. IR-A is predominantly expressed during prenatal life. It enhances the effects of IGF-II during embryogenesis and fetal development. It is also significantly expressed in adult tissues, especially in the brain. Conversely, IR-B is predominantly expressed in adult, well-differentiated tissues, including the liver, where it enhances the metabolic effects of insulin. Dysregulation of IR splicing in insulin target tissues may occur in patients with insulin resistance; however, its role in type 2 diabetes is unclear. IR-A is often aberrantly expressed in cancer cells, thus increasing their responsiveness to IGF-II and to insulin and explaining the cancer-promoting effect of hyperinsulinemia observed in obese and type 2 diabetic patients. Aberrant IR-A expression may favor cancer resistance to both conventional and targeted therapies by a variety of mechanisms. Finally, IR isoforms form heterodimers, IR-A/IR-B, and hybrid IR/IGF-IR receptors (HR-A and HR-B). The functional characteristics of such hybrid receptors and their role in physiology, in diabetes, and in malignant cells are not yet fully understood. These receptors seem to enhance cell responsiveness to IGFs.
Collapse
Affiliation(s)
- Antonino Belfiore
- Endocrinology, Department of Clinical and Experimental Medicine, University of Catania, Ospedale Garibaldi-Nesima, 95122 Catania, Italy.
| | | | | | | | | |
Collapse
|
188
|
Rafacho A, Giozzet V, Boschero A, Abrantes J, Cestari T, Carneiro E, Bosqueiro J. Reduced pancreatic β-cell mass is associated with decreased FoxO1 and Erk1/2 protein phosphorylation in low-protein malnourished rats. Braz J Med Biol Res 2009; 42:935-41. [DOI: 10.1590/s0100-879x2009001000010] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2009] [Accepted: 08/10/2009] [Indexed: 02/01/2023] Open
|
189
|
Li J, Johnson JD. MATHEMATICAL MODELS OF SUBCUTANEOUS INJECTION OF INSULIN ANALOGUES: A MINI-REVIEW. DISCRETE AND CONTINUOUS DYNAMICAL SYSTEMS. SERIES B 2009; 12:401-414. [PMID: 21572588 PMCID: PMC3093671 DOI: 10.3934/dcdsb.2009.12.401] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
In the last three decades, several models relevant to the subcutaneous injection of insulin analogues have appeared in the literature. Most of them model the absorption of insulin analogues in the injection depot and then compute the plasma insulin concentration. The most recent systemic models directly simulate the plasma insulin dynamics. These models have been and/or can be applied to the technology of the insulin pump or to the coming closed-loop systems, also known as the artificial pancreas. In this paper, we selectively review these models in detail and at point out that these models provide key building blocks for some important endeavors into physiological questions of insulin secretion and action. For example, it is not clear at this time whether or not picomolar doses of insulin are found near the islets and there is no experimental method to assess this in vivo. This is of interest because picomolar concentrations of insulin have been found to be effective at blocking beta-cell death and increasing beta-cell growth in recent cell culture experiments.
Collapse
Affiliation(s)
- Jiaxu Li
- Department of Mathematics, University of Louisville, Louisville, KY 40292, USA
| | - James D. Johnson
- Department of Cellular and Physiological Sciences; Department of Surgery, University of British Columbia, Vancouver, BC, Canada, V6T 1Z3
| |
Collapse
|
190
|
Abstract
The β-cells of the pancreas are responsible for insulin production and their destruction results in type I diabetes. β-cell maintenance, growth and regenerative repair is thought to occur predominately, if not exclusively, through the replication of existing β-cells, not via an adult stem cell. It was recently found that all β-cells contribute equally to islet growth and maintenance. The fact that all β-cells replicate homogeneously makes it possible to set up straightforward screens for factors that increase β-cell replication either In vitro or in vivo. It is possible that a circulating factor may be capable of increasing β-cell replication or that intrinsic cell cycle regulators may affect β-cell growth. An improved understanding of the in vivo maintenance and growth of β-cells will facilitate efforts to expand β-cells In vitro and may lead to new treatments for diabetes.
Collapse
Affiliation(s)
- Kristen Brennand
- Department of Stem Cell and Regenerative Biology, HHMI and Harvard University, Harvard Stem Cell Institute, Cambridge, MA, USA
| | | |
Collapse
|
191
|
Surampudi PN, John-Kalarickal J, Fonseca VA. Emerging concepts in the pathophysiology of type 2 diabetes mellitus. ACTA ACUST UNITED AC 2009; 76:216-26. [PMID: 19421965 DOI: 10.1002/msj.20113] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Type 2 diabetes mellitus is a multifactorial metabolic disorder. It is characterized by chronic hyperglycemia, insulin resistance, and a relative insulin secretion defect. The prevalence of type 2 diabetes mellitus has risen worldwide in large part because of an increase in obesity and sedentary lifestyles. The underlying pathophysiology and complications of type 2 diabetes mellitus are still being elucidated. Recent advances in diabetes research have helped us to gain a better understanding about insulin resistance and insulin secretion defects. The evolving understanding about the influence of the incretin effect, insulin signal transduction, adipose tissue, intra-islet cell communication, and inflammation is changing the way in which we view type 2 diabetes mellitus. This new understanding will eventually provide us with new treatment approaches to help patients who have type 2 diabetes mellitus. This article gives a review of the current and emerging concepts of the pathophysiology of type 2 diabetes mellitus.
Collapse
|
192
|
Annicotte JS, Blanchet E, Chavey C, Iankova I, Costes S, Assou S, Teyssier J, Dalle S, Sardet C, Fajas L. The CDK4-pRB-E2F1 pathway controls insulin secretion. Nat Cell Biol 2009; 11:1017-23. [PMID: 19597485 DOI: 10.1038/ncb1915] [Citation(s) in RCA: 108] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2008] [Accepted: 04/28/2009] [Indexed: 12/20/2022]
Abstract
CDK4-pRB-E2F1 cell-cycle regulators are robustly expressed in non-proliferating beta cells, suggesting that besides the control of beta-cell number the CDK4-pRB-E2F1 pathway has a role in beta-cell function. We show here that E2F1 directly regulates expression of Kir6.2, which is a key component of the K(ATP) channel involved in the regulation of glucose-induced insulin secretion. We demonstrate, through chromatin immunoprecipitation analysis from tissues, that Kir6.2 expression is regulated at the promoter level by the CDK4-pRB-E2F1 pathway. Consistently, inhibition of CDK4, or genetic inactivation of E2F1, results in decreased expression of Kir6.2, impaired insulin secretion and glucose intolerance in mice. Furthermore we show that rescue of Kir6.2 expression restores insulin secretion in E2f1(-/-) beta cells. Finally, we demonstrate that CDK4 is activated by glucose through the insulin pathway, ultimately resulting in E2F1 activation and, consequently, increased expression of Kir6.2. In summary we provide evidence that the CDK4-pRB-E2F1 regulatory pathway is involved in glucose homeostasis, defining a new link between cell proliferation and metabolism.
Collapse
|
193
|
Martens GA, Pipeleers D. Glucose, regulator of survival and phenotype of pancreatic beta cells. VITAMINS AND HORMONES 2009; 80:507-39. [PMID: 19251048 DOI: 10.1016/s0083-6729(08)00617-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The key role of glucose in regulating insulin release by the pancreatic beta cell population is not only dependent on acute stimulus-secretion coupling mechanisms but also on more long-term influences on beta cell survival and phenotype. Glucose serves as a major survival factor for beta cells via at least three actions: it prevents an oxidative redox state, it suppresses a mitochondrial apoptotic program that is triggered at reduced mitochondrial metabolic activity and it induces genes needed for the cellular responsiveness to glucose and to growth factors. Glucose-regulated pathways may link protein synthetic and proliferative activities, making glucose a permissive factor for beta cell proliferation, in check with metabolic needs. Conditions of inadequate glucose metabolism in beta cells are not only leading to deregulation of acute secretory responses but should also be considered as causes for increased apoptosis and reduced formation of beta cells, and loss of their normal differentiated state.
Collapse
|
194
|
Coordinated regulation by Shp2 tyrosine phosphatase of signaling events controlling insulin biosynthesis in pancreatic beta-cells. Proc Natl Acad Sci U S A 2009; 106:7531-6. [PMID: 19380737 DOI: 10.1073/pnas.0811715106] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Intracellular signaling by which pancreatic beta-cells synthesize and secrete insulin in control of glucose homeostasis is not fully understood. Here we show that Shp2, a cytoplasmic tyrosine phosphatase possessing 2 SH2 domains, coordinates signaling events required for insulin biosynthesis in beta-cells. Mice with conditional ablation of the Shp2/Ptpn11 gene in the pancreas exhibited defective glucose-stimulated insulin secretion and impaired glucose tolerance. Consistently, siRNA-mediated Shp2-knockdown in rat insulinoma INS-1 832/13 cells resulted in decreased insulin production and secretion despite an increase in cellular ATP. Shp2 modulates the strength of signals flowing through Akt/FoxO1 and Erk pathways, culminating in control of Pdx1 expression and activity on Ins1 and Ins2 promoters, and forced Pdx1 expression rescued insulin production in Shp2-knockdown beta-cells. Therefore, Shp2 acts as a signal coordinator in beta-cells, orchestrating multiple pathways controlling insulin biosynthesis to maintain glucose homeostasis.
Collapse
|
195
|
Rafacho A, Cestari TM, Taboga SR, Boschero AC, Bosqueiro JR. High doses of dexamethasone induce increased beta-cell proliferation in pancreatic rat islets. Am J Physiol Endocrinol Metab 2009; 296:E681-9. [PMID: 19158320 DOI: 10.1152/ajpendo.90931.2008] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Activation of insulin signaling and cell cycle intermediates is required for adult beta-cell proliferation. Here, we report a model to study beta-cell proliferation in living rats by administering three different doses of dexamethasone (0.1, 0.5, and 1.0 mg/kg ip, DEX 0.1, DEX 0.5, and DEX 1.0, respectively) for 5 days. Insulin sensitivity, insulin secretion, and histomorphometric data were investigated. Western blotting was used to analyze the levels of proteins related to the control of beta-cell growth. DEX 1.0 rats, which present moderate hyperglycemia and marked hyperinsulinemia, exhibited a 5.1-fold increase in beta-cell proliferation and an increase (17%) in beta-cell size, with significant increase in beta-cell mass, compared with control rats. The hyperinsulinemic but euglycemic DEX 0.5 rats also showed a significant 3.6-fold increase in beta-cell proliferation. However, DEX 0.1 rats, which exhibited the lowest degree of insulin resistance, compensate for insulin demand by improving only islet function. Activation of the insulin receptor substrate 2/phosphatidylinositol 3-kinase/serine-threonine kinase/ribosomal protein S6 kinase pathway, as well as protein retinoblastoma in islets from DEX 1.0 and DEX 0.5, but not in DEX 0.1, rats was also observed. Therefore, increasing doses of dexamethasone induce three different degrees of insulin requirement in living rats, serving as a model to investigate compensatory beta-cell alterations. Augmented beta-cell mass involves beta-cell hyperplasia and, to a lower extent, beta-cell hypertrophy. We suggest that alterations in circulating insulin and, to a lesser extent, glucose levels could be the major stimuli for beta-cell proliferation in the dexamethasone-induced insulin resistance.
Collapse
Affiliation(s)
- Alex Rafacho
- Department of Physiology and Biophysics, Institute of Biology, University of Campinas, Rua Monteiro Lobato, s/n. Cidade Universitária, Campinas, São Paulo, Brazil, 13083-970.
| | | | | | | | | |
Collapse
|
196
|
Escribano O, Guillén C, Nevado C, Gómez-Hernández A, Kahn CR, Benito M. Beta-Cell hyperplasia induced by hepatic insulin resistance: role of a liver-pancreas endocrine axis through insulin receptor A isoform. Diabetes 2009; 58:820-8. [PMID: 19136656 PMCID: PMC2661585 DOI: 10.2337/db08-0551] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Type 2 diabetes results from a combination of insulin resistance and impaired insulin secretion. To directly address the effects of hepatic insulin resistance in adult animals, we developed an inducible liver-specific insulin receptor knockout mouse (iLIRKO). RESEARCH DESIGN AND METHODS Using this approach, we were able to induce variable insulin receptor (IR) deficiency in a tissue-specific manner (liver mosaicism). RESULTS iLIRKO mice presented progressive hepatic and extrahepatic insulin resistance without liver dysfunction. Initially, iLIRKO mice displayed hyperinsulinemia and increased beta-cell mass, the extent of which was proportional to the deletion of hepatic IR. Our studies of iLIRKO suggest a cause-and-effect relationship between progressive insulin resistance and the fold increase of plasma insulin levels and beta-cell mass. Ultimately, the beta-cells failed to secrete sufficient insulin, leading to uncontrolled diabetes. We observed that hepatic IGF-1 expression was enhanced in iLIRKO mice, resulting in an increase of circulating IGF-1. Concurrently, the IR-A isoform was upregulated in hyperplastic beta-cells of iLIRKO mice and IGF-1-induced proliferation was higher than in the controls. In mouse beta-cell lines, IR-A, but not IR-B, conferred a proliferative capacity in response to insulin or IGF-1, providing a potential explanation for the beta-cell hyperplasia induced by liver insulin resistance in iLIRKO mice. CONCLUSIONS Our studies of iLIRKO mice suggest a liver-pancreas endocrine axis in which IGF-1 functions as a liver-derived growth factor to promote compensatory pancreatic islet hyperplasia through IR-A.
Collapse
Affiliation(s)
- Oscar Escribano
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, Madrid, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain; and
| | - Carlos Guillén
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, Madrid, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain; and
| | - Carmen Nevado
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, Madrid, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain; and
| | - Almudena Gómez-Hernández
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, Madrid, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain; and
| | - C. Ronald Kahn
- Research Division, Joslin Diabetes Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Manuel Benito
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, Madrid, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain; and
- Corresponding author: Manuel Benito,
| |
Collapse
|
197
|
Peroxisome proliferator-activated receptor gamma activation restores islet function in diabetic mice through reduction of endoplasmic reticulum stress and maintenance of euchromatin structure. Mol Cell Biol 2009; 29:2053-67. [PMID: 19237535 DOI: 10.1128/mcb.01179-08] [Citation(s) in RCA: 122] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The nuclear receptor peroxisome proliferator-activated receptor gamma (PPAR-gamma) is an important target in diabetes therapy, but its direct role, if any, in the restoration of islet function has remained controversial. To identify potential molecular mechanisms of PPAR-gamma in the islet, we treated diabetic or glucose-intolerant mice with the PPAR-gamma agonist pioglitazone or with a control. Treated mice exhibited significantly improved glycemic control, corresponding to increased serum insulin and enhanced glucose-stimulated insulin release and Ca(2+) responses from isolated islets in vitro. This improved islet function was at least partially attributed to significant upregulation of the islet genes Irs1, SERCA, Ins1/2, and Glut2 in treated animals. The restoration of the Ins1/2 and Glut2 genes corresponded to a two- to threefold increase in the euchromatin marker histone H3 dimethyl-Lys4 at their respective promoters and was coincident with increased nuclear occupancy of the islet methyltransferase Set7/9. Analysis of diabetic islets in vitro suggested that these effects resulting from the presence of the PPAR-gamma agonist may be secondary to improvements in endoplasmic reticulum stress. Consistent with this possibility, incubation of thapsigargin-treated INS-1 beta cells with the PPAR-gamma agonist resulted in the reduction of endoplasmic reticulum stress and restoration of Pdx1 protein levels and Set7/9 nuclear occupancy. We conclude that PPAR-gamma agonists exert a direct effect in diabetic islets to reduce endoplasmic reticulum stress and enhance Pdx1 levels, leading to favorable alterations of the islet gene chromatin architecture.
Collapse
|
198
|
Sachdeva MM, Stoffers DA. Minireview: Meeting the demand for insulin: molecular mechanisms of adaptive postnatal beta-cell mass expansion. Mol Endocrinol 2009; 23:747-58. [PMID: 19196831 DOI: 10.1210/me.2008-0400] [Citation(s) in RCA: 121] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Type 2 diabetes results from pancreatic ss-cell failure in the setting of insulin resistance. This model of disease progression has received recent support from the results of genome-wide association studies that identify genes potentially regulating ss-cell growth and function as type 2 diabetes susceptibility loci. Normal ss-cell compensation for an increased insulin demand includes both enhanced insulin-secretory capacity and an expansion of morphological ss-cell mass, due largely to changes in the balance between ss-cell proliferation and apoptosis. Recent years have brought significant progress in the understanding of both extrinsic signals stimulating ss-cell growth as well as mediators intrinsic to the ss-cell that regulate the compensatory response. Here, we review the current knowledge of mechanisms underlying adaptive expansion of ss-cell mass, focusing on lessons learned from experimental models of physiologically occurring insulin-resistant states including diet-induced obesity and pregnancy, and highlighting the potential importance of interorgan cross talk. The identification of critical mediators of islet compensation may direct the development of future therapeutic strategies to enhance the response of ss-cells to insulin resistance.
Collapse
Affiliation(s)
- Mira M Sachdeva
- Department of Medicine, Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania School of Medicine, Philadelphia, 19104, USA
| | | |
Collapse
|
199
|
Abstract
Type 1 and type 2 diabetes mellitus together are predicted to affect over 300 million people worldwide by the year 2020. A relative or absolute paucity of functional β-cells is a central feature of both types of disease, and identifying the pathways that mediate the embryonic origin of new β-cells and mechanisms that underlie the proliferation of existing β-cells are major efforts in the fields of developmental and islet biology. A poor secretory response of existing β-cells to nutrients and hormones and the defects in hormone processing also contribute to the hyperglycemia observed in type 2 diabetes and has prompted studies aimed at enhancing β-cell function. The factors that contribute to a greater susceptibility in aging individuals to develop diabetes is currently unclear and may be linked to a poor turnover of β-cells and/or enhanced susceptibility of β-cells to apoptosis. This review is an update on the recent work in the areas of islet/β-cell regeneration and hormone processing that are relevant to the pathophysiology of the endocrine pancreas in type 1, type 2 and obesity-associated diabetes.
Collapse
Affiliation(s)
- Anke Assmann
- Research Division, Joslin Diabetes Center and Department of Medicine, Harvard Medical School, Boston, MA 02215, USA
| | | | | |
Collapse
|
200
|
Lee YC, Nielsen JH. Regulation of beta cell replication. Mol Cell Endocrinol 2009; 297:18-27. [PMID: 18824066 DOI: 10.1016/j.mce.2008.08.033] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2008] [Revised: 08/24/2008] [Accepted: 08/24/2008] [Indexed: 10/21/2022]
Abstract
Beta cell mass, at any given time, is governed by cell differentiation, neogenesis, increased or decreased cell size (cell hypertrophy or atrophy), cell death (apoptosis), and beta cell proliferation. Nutrients, hormones and growth factors coupled with their signalling intermediates have been suggested to play a role in beta cell mass regulation. In addition, genetic mouse model studies have indicated that cyclins and cyclin-dependent kinases that determine cell cycle progression are involved in beta cell replication, and more recently, menin in association with cyclin-dependent kinase inhibitors has been demonstrated to be important in beta cell growth. In this review, we consider and highlight some aspects of cell cycle regulation in relation to beta cell replication. The role of cell cycle regulation in beta cell replication is mostly from studies in rodent models, but whether these findings can be extended to human beta cells remains to be shown.
Collapse
Affiliation(s)
- Ying C Lee
- Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, Building 6.5, DK-2200 Copenhagen N., Denmark
| | | |
Collapse
|