101
|
Abstract
Glucose homeostasis is precisely regulated by glucagon and insulin, which are released by pancreatic α- and β-cells, respectively. While β-cells have been the focus of intense research, less is known about α-cell function and the actions of glucagon. In recent years, the study of this endocrine cell type has experienced a renewed drive. The present review contains a summary of established concepts as well as new information about the regulation of α-cells by glucose, amino acids, fatty acids and other nutrients, focusing especially on glucagon release, glucagon synthesis and α-cell survival. We have also discussed the role of glucagon in glucose homeostasis and in energy and lipid metabolism as well as its potential as a modulator of food intake and body weight. In addition to the well-established action on the liver, we discuss the effects of glucagon in other organs, where the glucagon receptor is expressed. These tissues include the heart, kidneys, adipose tissue, brain, small intestine and the gustatory epithelium. Alterations in α-cell function and abnormal glucagon concentrations are present in diabetes and are thought to aggravate the hyperglycaemic state of diabetic patients. In this respect, several experimental approaches in diabetic models have shown important beneficial results in improving hyperglycaemia after the modulation of glucagon secretion or action. Moreover, glucagon receptor agonism has also been used as a therapeutic strategy to treat obesity.
Collapse
|
102
|
Strömstedt AA, Felth J, Bohlin L. Bioassays in natural product research - strategies and methods in the search for anti-inflammatory and antimicrobial activity. PHYTOCHEMICAL ANALYSIS : PCA 2014; 25:13-28. [PMID: 24019222 DOI: 10.1002/pca.2468] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Revised: 06/24/2013] [Accepted: 06/24/2013] [Indexed: 06/02/2023]
Abstract
INTRODUCTION Identifying bioactive molecules from complex biomasses requires careful selection and execution of relevant bioassays in the various stages of the discovery process of potential leads and targets. OBJECTIVE The aim of this review is to share our long-term experience in bioassay-guided isolation, and mechanistic studies, of bioactive compounds from different organisms in nature with emphasis on anti-inflammatory and antimicrobial activity. METHODS In the search for anti-inflammatory activity, in vivo and in vitro model combinations with enzymes and cells involved in the inflammatory process have been used, such as cyclooxygenases, human neutrophils and human cancer cell lines. Methods concerning adsorption and perforation of bacteria, fungi, human cells and model membranes, have been developed and optimised, with emphasis on antimicrobial peptides and their interaction with the membrane target, in particular their ability to distinguish host from pathogen. RESULTS A long-term research has provided experience of selection and combination of bioassay models, which has led to an increased understanding of ethnopharmacological and ecological observations, together with in-depth knowledge of mode of action of isolated compounds. CONCLUSION A more multidisciplinary approach and a higher degree of fundamental research in development of bioassays are often necessary to identify and to fully understand the mode of action of bioactive molecules with novel structure-activity relationships from natural sources.
Collapse
Affiliation(s)
- Adam A Strömstedt
- Division of Pharmacognosy, Department of Medicinal Chemistry, Biomedical Center, Uppsala University, Box 574, 751 23, Uppsala, Sweden
| | | | | |
Collapse
|
103
|
Abstract
Glucagon secreted by pancreatic α-cells is the major hyperglycemic hormone correcting acute hypoglycaemia (glucose counterregulation). In diabetes the glucagon response to hypoglycaemia becomes compromised and chronic hyperglucagonemia appears. There is increasing awareness that glucagon excess may underlie important manifestations of diabetes. However opinions differ widely how glucose controls glucagon secretion. The autonomous nervous system plays an important role in the glucagon response to hypoglycaemia. But it is clear that glucose controls glucagon secretion also by mechanisms involving direct effects on α-cells or indirect effects via paracrine factors released from non-α-cells within the pancreatic islets. The present review discusses these mechanisms and argues that different regulatory processes are involved in a glucose concentration-dependent manner. Direct glucose effects on the α-cell and autocrine mechanisms are probably most significant for the glucagon response to hypoglycaemia. During hyperglycaemia, when secretion from β- and δ-cells is stimulated, paracrine inhibitory factors generate pulsatile glucagon release in opposite phase to pulsatile release of insulin and somatostatin. High concentrations of glucose have also stimulatory effects on glucagon secretion that tend to balance and even exceed the inhibitory influence. The latter actions might underlie the paradoxical hyperglucagonemia that aggravates hyperglycaemia in persons with diabetes.
Collapse
Affiliation(s)
- Erik Gylfe
- Department of Medical Cell Biology, Uppsala University, BMC Box 571, SE-751 23, Uppsala, Sweden.
| | - Patrick Gilon
- Pôle d'Endocrinologie, Diabète et Nutrition, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
104
|
Zhang Q, Ramracheya R, Lahmann C, Tarasov A, Bengtsson M, Braha O, Braun M, Brereton M, Collins S, Galvanovskis J, Gonzalez A, Groschner LN, Rorsman NJG, Salehi A, Travers ME, Walker JN, Gloyn AL, Gribble F, Johnson PRV, Reimann F, Ashcroft FM, Rorsman P. Role of KATP channels in glucose-regulated glucagon secretion and impaired counterregulation in type 2 diabetes. Cell Metab 2013; 18:871-82. [PMID: 24315372 PMCID: PMC3851686 DOI: 10.1016/j.cmet.2013.10.014] [Citation(s) in RCA: 166] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Revised: 07/05/2013] [Accepted: 10/31/2013] [Indexed: 01/03/2023]
Abstract
Glucagon, secreted by pancreatic islet α cells, is the principal hyperglycemic hormone. In diabetes, glucagon secretion is not suppressed at high glucose, exacerbating the consequences of insufficient insulin secretion, and is inadequate at low glucose, potentially leading to fatal hypoglycemia. The causal mechanisms remain unknown. Here we show that α cell KATP-channel activity is very low under hypoglycemic conditions and that hyperglycemia, via elevated intracellular ATP/ADP, leads to complete inhibition. This produces membrane depolarization and voltage-dependent inactivation of the Na(+) channels involved in action potential firing that, via reduced action potential height and Ca(2+) entry, suppresses glucagon secretion. Maneuvers that increase KATP channel activity, such as metabolic inhibition, mimic the glucagon secretory defects associated with diabetes. Low concentrations of the KATP channel blocker tolbutamide partially restore glucose-regulated glucagon secretion in islets from type 2 diabetic organ donors. These data suggest that impaired metabolic control of the KATP channels underlies the defective glucose regulation of glucagon secretion in type 2 diabetes.
Collapse
Affiliation(s)
- Quan Zhang
- Oxford Centre for Diabetes, Endocrinology, and Metabolism, University of Oxford, Churchill Hospital, Oxford OX3 7LJ, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
105
|
Cura AJ, Carruthers A. Role of monosaccharide transport proteins in carbohydrate assimilation, distribution, metabolism, and homeostasis. Compr Physiol 2013; 2:863-914. [PMID: 22943001 DOI: 10.1002/cphy.c110024] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The facilitated diffusion of glucose, galactose, fructose, urate, myoinositol, and dehydroascorbicacid in mammals is catalyzed by a family of 14 monosaccharide transport proteins called GLUTs. These transporters may be divided into three classes according to sequence similarity and function/substrate specificity. GLUT1 appears to be highly expressed in glycolytically active cells and has been coopted in vitamin C auxotrophs to maintain the redox state of the blood through transport of dehydroascorbate. Several GLUTs are definitive glucose/galactose transporters, GLUT2 and GLUT5 are physiologically important fructose transporters, GLUT9 appears to be a urate transporter while GLUT13 is a proton/myoinositol cotransporter. The physiologic substrates of some GLUTs remain to be established. The GLUTs are expressed in a tissue specific manner where affinity, specificity, and capacity for substrate transport are paramount for tissue function. Although great strides have been made in characterizing GLUT-catalyzed monosaccharide transport and mapping GLUT membrane topography and determinants of substrate specificity, a unifying model for GLUT structure and function remains elusive. The GLUTs play a major role in carbohydrate homeostasis and the redistribution of sugar-derived carbons among the various organ systems. This is accomplished through a multiplicity of GLUT-dependent glucose sensing and effector mechanisms that regulate monosaccharide ingestion, absorption,distribution, cellular transport and metabolism, and recovery/retention. Glucose transport and metabolism have coevolved in mammals to support cerebral glucose utilization.
Collapse
Affiliation(s)
- Anthony J Cura
- Department of Biochemistry & Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | | |
Collapse
|
106
|
Gylfe E. Comment on: Allister et al. UCP2 regulates the glucagon response to fasting and starvation. Diabetes 2013;62:1623-1633. Diabetes 2013; 62:e11. [PMID: 23881203 PMCID: PMC3717851 DOI: 10.2337/db13-0397] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
107
|
Vieira E, Marroquí L, Figueroa ALC, Merino B, Fernandez-Ruiz R, Nadal A, Burris TP, Gomis R, Quesada I. Involvement of the clock gene Rev-erb alpha in the regulation of glucagon secretion in pancreatic alpha-cells. PLoS One 2013; 8:e69939. [PMID: 23936124 PMCID: PMC3723646 DOI: 10.1371/journal.pone.0069939] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Accepted: 06/13/2013] [Indexed: 11/19/2022] Open
Abstract
Disruption of pancreatic clock genes impairs pancreatic beta-cell function, leading to the onset of diabetes. Despite the importance of pancreatic alpha-cells in the regulation of glucose homeostasis and in diabetes pathophysiology, nothing is known about the role of clock genes in these cells. Here, we identify the clock gene Rev-erb alpha as a new intracellular regulator of glucagon secretion. Rev-erb alpha down-regulation by siRNA (60–70% inhibition) in alphaTC1-9 cells inhibited low-glucose induced glucagon secretion (p<0.05) and led to a decrease in key genes of the exocytotic machinery. The Rev-erb alpha agonist GSK4112 increased glucagon secretion (1.6 fold) and intracellular calcium signals in alphaTC1-9 cells and mouse primary alpha-cells, whereas the Rev-erb alpha antagonist SR8278 produced the opposite effect. At 0.5 mM glucose, alphaTC1-9 cells exhibited intrinsic circadian Rev-erb alpha expression oscillations that were inhibited by 11 mM glucose. In mouse primary alpha-cells, glucose induced similar effects (p<0.001). High glucose inhibited key genes controlled by AMPK such as Nampt, Sirt1 and PGC-1 alpha in alphaTC1-9 cells (p<0.05). AMPK activation by metformin completely reversed the inhibitory effect of glucose on Nampt-Sirt1-PGC-1 alpha and Rev-erb alpha. Nampt inhibition decreased Sirt1, PGC-1 alpha and Rev-erb alpha mRNA expression (p<0.01) and glucagon release (p<0.05). These findings identify Rev-erb alpha as a new intracellular regulator of glucagon secretion via AMPK/Nampt/Sirt1 pathway.
Collapse
MESH Headings
- AMP-Activated Protein Kinases/genetics
- AMP-Activated Protein Kinases/metabolism
- Animals
- Cell Line
- Circadian Rhythm/genetics
- Cytokines/genetics
- Cytokines/metabolism
- Gene Expression Regulation
- Glucagon/genetics
- Glucagon/metabolism
- Glucagon-Secreting Cells/cytology
- Glucagon-Secreting Cells/drug effects
- Glucagon-Secreting Cells/metabolism
- Glucose/metabolism
- Glucose/pharmacology
- Glycine/analogs & derivatives
- Glycine/pharmacology
- Isoquinolines/pharmacology
- Metformin/pharmacology
- Mice
- Nicotinamide Phosphoribosyltransferase/genetics
- Nicotinamide Phosphoribosyltransferase/metabolism
- Nuclear Receptor Subfamily 1, Group D, Member 1/agonists
- Nuclear Receptor Subfamily 1, Group D, Member 1/antagonists & inhibitors
- Nuclear Receptor Subfamily 1, Group D, Member 1/genetics
- Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Signal Transduction
- Sirtuin 1/genetics
- Sirtuin 1/metabolism
- Thiophenes/pharmacology
Collapse
Affiliation(s)
- Elaine Vieira
- Instituto de Bioingeniería, Universidad Miguel Hernandez de Elche, Elche, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Barcelona, Spain
- * E-mail: (EV); (IQ)
| | - Laura Marroquí
- Instituto de Bioingeniería, Universidad Miguel Hernandez de Elche, Elche, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Barcelona, Spain
| | - Ana Lucia C. Figueroa
- Diabetes and Obesity Laboratory, Institut d'investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Beatriz Merino
- Instituto de Bioingeniería, Universidad Miguel Hernandez de Elche, Elche, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Barcelona, Spain
| | - Rebeca Fernandez-Ruiz
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Barcelona, Spain
- Diabetes and Obesity Laboratory, Institut d'investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Angel Nadal
- Instituto de Bioingeniería, Universidad Miguel Hernandez de Elche, Elche, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Barcelona, Spain
| | - Thomas P. Burris
- Department of Molecular Therapeutics, The Scripps Research Institute, Jupiter, Florida, United States of America
| | - Ramon Gomis
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Barcelona, Spain
- Diabetes and Obesity Laboratory, Institut d'investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Endocrinology and Diabetes Unit, Hospital Clinic, Universitat de Barcelona, Barcelona, Spain
| | - Ivan Quesada
- Instituto de Bioingeniería, Universidad Miguel Hernandez de Elche, Elche, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Barcelona, Spain
- * E-mail: (EV); (IQ)
| |
Collapse
|
108
|
Gosmain Y, Masson MH, Philippe J. Glucagon: the renewal of an old hormone in the pathophysiology of diabetes. J Diabetes 2013; 5:102-9. [PMID: 23302052 DOI: 10.1111/1753-0407.12022] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Accepted: 12/14/2012] [Indexed: 12/24/2022] Open
Abstract
Type 2 diabetes (T2D) is one of the most common diseases, affecting 5-10% of the population in most countries; the progression of its prevalence has been constant over the past 50 years in all countries worldwide, creating a major public health problem in terms of disease management and financial burden. Although the pathophysiology of T2D has been attributed for decades to insulin resistance and decreased insulin secretion, particularly in response to glucose, the contributing role of glucagon in hyperglycemia has been highlighted since the early 1970s by demonstrating its glycogenolytic, gluconeogenic and ketogenic properties. More recently, the importance of glucagon in diabetes has been highlighted in a model of streptozotocin-induced diabetic mice becoming euglycemic in the absence of glucagon receptors and without insulin treatment. Understanding the dysregulation of α-cells in diabetes will be critical to better define the pathophysiology of diabetes and develop new antidiabetic treatment.
Collapse
Affiliation(s)
- Yvan Gosmain
- Service of Endocrinology, Diabetes, Hypertension and Nutrition, University Hospital Geneva, Geneva, Switzerland
| | | | | |
Collapse
|
109
|
Cuesta S, Kireev R, García C, Rancan L, Vara E, Tresguerres JAF. Melatonin can improve insulin resistance and aging-induced pancreas alterations in senescence-accelerated prone male mice (SAMP8). AGE (DORDRECHT, NETHERLANDS) 2013; 35:659-671. [PMID: 22411259 PMCID: PMC3636397 DOI: 10.1007/s11357-012-9397-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2011] [Accepted: 02/20/2012] [Indexed: 05/31/2023]
Abstract
The aim of the present study was to investigate the effect of aging on several parameters related to glucose homeostasis and insulin resistance in pancreas and how melatonin administration could affect these parameters. Pancreas samples were obtained from two types of male mice models: senescence-accelerated prone (SAMP8) and senescence-accelerated-resistant mice (SAMR1). Insulin levels in plasma were increased with aging in both SAMP8 and SAMR1 mice, whereas insulin content in pancreas was decreased with aging in SAMP8 and increased in SAMR1 mice. Expressions of glucagon and GLUT2 messenger RNAs (mRNAs) were increased with aging in SAMP8 mice, and no differences were observed in somatostatin and insulin mRNA expressions. Furthermore, aging decreased also the expressions of Pdx-1, FoxO 1, FoxO 3A and Sirt1 in pancreatic SAMP8 samples. Pdx-1 was decreased in SAMR1 mice, but no differences were observed in the rest of parameters on these mice strains. Treatment with melatonin was able to decrease plasma insulin levels and to increase its pancreatic content in SAMP8 mice. In SAMR1, insulin pancreatic content and plasma levels were decreased. HOMA-IR was decreased with melatonin treatment in both strains of animals. On the other hand, in SAMP8 mice, treatment decreased the expression of glucagon, GLUT2, somatostatin and insulin mRNA. Furthermore, it was also able to increase the expression of Sirt1, Pdx-1 and FoxO 3A. According to these results, aging is associated with significant alterations in the relative expression of pancreatic genes associated to glucose metabolism. This has been especially observed in SAMP8 mice. Melatonin administration was able to improve pancreatic function in old SAMP8 mice and to reduce HOMA-IR improving their insulin physiology and glucose metabolism.
Collapse
Affiliation(s)
- Sara Cuesta
- />Department of Physiology, Medical School, University Complutense of Madrid, Madrid, Spain
| | - Roman Kireev
- />Department of Physiology, Medical School, University Complutense of Madrid, Madrid, Spain
| | - Cruz García
- />Department of Biochemistry and Molecular Biology, Medical School, University Complutense of Madrid, Madrid, Spain
| | - Lisa Rancan
- />Department of Biochemistry and Molecular Biology, Medical School, University Complutense of Madrid, Madrid, Spain
| | - Elena Vara
- />Department of Biochemistry and Molecular Biology, Medical School, University Complutense of Madrid, Madrid, Spain
| | - Jesús A. F. Tresguerres
- />Department of Physiology, Medical School, University Complutense of Madrid, Madrid, Spain
- />Laboratory of Experimental Endocrinology, Department of Physiology, School of Medicine, Complutense University, Avda, Complutense s/n, 28040 Madrid, Spain
| |
Collapse
|
110
|
Cheng-Xue R, Gómez-Ruiz A, Antoine N, Noël LA, Chae HY, Ravier MA, Chimienti F, Schuit FC, Gilon P. Tolbutamide controls glucagon release from mouse islets differently than glucose: involvement of K(ATP) channels from both α-cells and δ-cells. Diabetes 2013; 62:1612-22. [PMID: 23382449 PMCID: PMC3636641 DOI: 10.2337/db12-0347] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We evaluated the role of ATP-sensitive K⁺ (K(ATP)) channels, somatostatin, and Zn²⁺ in the control of glucagon secretion from mouse islets. Switching from 1 to 7 mmol/L glucose inhibited glucagon release. Diazoxide did not reverse the glucagonostatic effect of glucose. Tolbutamide decreased glucagon secretion at 1 mmol/L glucose (G1) but stimulated it at 7 mmol/L glucose (G7). The reduced glucagon secretion produced by high concentrations of tolbutamide or diazoxide, or disruption of K(ATP) channels (Sur1(-/-) mice) at G1 could be inhibited further by G7. Removal of the somatostatin paracrine influence (Sst(-/-) mice or pretreatement with pertussis toxin) strongly increased glucagon release, did not prevent the glucagonostatic effect of G7, and unmasked a marked glucagonotropic effect of tolbutamide. Glucose inhibited glucagon release in the absence of functional K(ATP) channels and somatostatin signaling. Knockout of the Zn²⁺ transporter ZnT8 (ZnT8(-/-) mice) did not prevent the glucagonostatic effect of glucose. In conclusion, glucose can inhibit glucagon release independently of Zn²⁺, K(ATP) channels, and somatostatin. Closure of K(ATP) channels controls glucagon secretion by two mechanisms, a direct stimulation of α-cells and an indirect inhibition via somatostatin released from δ-cells. The net effect on glucagon release results from a balance between both effects.
Collapse
Affiliation(s)
- Rui Cheng-Xue
- Pôle d’Endocrinologie, Diabète et Nutrition, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Ana Gómez-Ruiz
- Pôle d’Endocrinologie, Diabète et Nutrition, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Nancy Antoine
- Pôle d’Endocrinologie, Diabète et Nutrition, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Laura A. Noël
- Pôle d’Endocrinologie, Diabète et Nutrition, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Hee-Young Chae
- Pôle d’Endocrinologie, Diabète et Nutrition, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Magalie A. Ravier
- Institut de Génomique Fonctionnelle, CNRS UMR-5203, INSERM U661, Universités de Montpellier 1 et 2, Montpellier, France
| | | | - Frans C. Schuit
- Gene Expression Unit, Department of Molecular and Cellular Medicine, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Patrick Gilon
- Pôle d’Endocrinologie, Diabète et Nutrition, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
- Corresponding author: Patrick Gilon,
| |
Collapse
|
111
|
Affiliation(s)
- Erik Gylfe
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
112
|
Gaisano HY, Macdonald PE, Vranic M. Glucagon secretion and signaling in the development of diabetes. Front Physiol 2012; 3:349. [PMID: 22969729 PMCID: PMC3432929 DOI: 10.3389/fphys.2012.00349] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Accepted: 08/10/2012] [Indexed: 12/19/2022] Open
Abstract
Normal release of glucagon from pancreatic islet α-cells promotes glucose mobilization, which counteracts the hypoglycemic actions of insulin, thereby ensuring glucose homeostasis. In treatment of diabetes aimed at rigorously reducing hyperglycemia to avoid chronic complications, the resulting hypoglycemia triggering glucagon release from α-cells is frequently impaired, with ensuing hypoglycemic complications. This review integrates the physiology of glucagon secretion regulating glucose homeostasis in vivo to single α-cell signaling, and how both become perturbed in diabetes. α-cells within the social milieu of the islet micro-organ are regulated not only by intrinsic signaling events but also by paracrine regulation, particularly by adjacent insulin-secreting β-cells and somatostatin-secreting δ-cells. We discuss the intrinsic α-cell signaling events, including glucose sensing and ion channel regulation leading to glucagon secretion. We then discuss the complex crosstalk between the islet cells and the breakdown of this crosstalk in diabetes contributing to the dysregulated glucagon secretion. Whereas, there are many secretory products released by β- and δ-cells that become deficient or excess in diabetes, we discuss the major ones, including the better known insulin and lesser known somatostatin, which act as putative paracrine on/off switches that very finely regulate α-cell secretory responses in health and diabetes. Of note in several type 1 diabetes (T1D) rodent models, blockade of excess somatostatin actions on α-cell could normalize glucagon secretion sufficient to attain normoglycemia in response to hypoglycemic assaults. There has been slow progress in fully elucidating the pathophysiology of the α-cell in diabetes because of the small number of α-cells within an islet and the islet mass becomes severely reduced and inflamed in diabetes. These limitations are just now being surmounted by new approaches.
Collapse
Affiliation(s)
- Herbert Y Gaisano
- Departments of Medicine and Physiology, University of Toronto Toronto, ON, Canada
| | | | | |
Collapse
|
113
|
Tian G, Sågetorp J, Xu Y, Shuai H, Degerman E, Tengholm A. Role of phosphodiesterases in the shaping of sub-plasma-membrane cAMP oscillations and pulsatile insulin secretion. J Cell Sci 2012; 125:5084-95. [PMID: 22946044 DOI: 10.1242/jcs.107201] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Specificity and versatility in cyclic AMP (cAMP) signalling are governed by the spatial localisation and temporal dynamics of the signal. Phosphodiesterases (PDEs) are important for shaping cAMP signals by hydrolyzing the nucleotide. In pancreatic β-cells, glucose triggers sub-plasma-membrane cAMP oscillations, which are important for insulin secretion, but the mechanisms underlying the oscillations are poorly understood. Here, we investigated the role of different PDEs in the generation of cAMP oscillations by monitoring the concentration of cAMP in the sub-plasma-membrane space ([cAMP](pm)) with ratiometric evanescent wave microscopy in MIN6 cells or mouse pancreatic β-cells expressing a fluorescent translocation biosensor. The general PDE inhibitor IBMX increased [cAMP](pm), and whereas oscillations were frequently observed at 50 µM IBMX, 300 µM-1 mM of the inhibitor caused a stable increase in [cAMP](pm). The [cAMP](pm) was nevertheless markedly suppressed by the adenylyl cyclase inhibitor 2',5'-dideoxyadenosine, indicating IBMX-insensitive cAMP degradation. Among IBMX-sensitive PDEs, PDE3 was most important for maintaining a low basal level of [cAMP](pm) in unstimulated cells. After glucose induction of [cAMP](pm) oscillations, inhibitors of PDE1, PDE3 and PDE4 inhibitors the average cAMP level, often without disturbing the [cAMP](pm) rhythmicity. Knockdown of the IBMX-insensitive PDE8B by shRNA in MIN6 cells increased the basal level of [cAMP](pm) and prevented the [cAMP](pm)-lowering effect of 2',5'-dideoxyadenosine after exposure to IBMX. Moreover, PDE8B-knockdown cells showed reduced glucose-induced [cAMP](pm) oscillations and loss of the normal pulsatile pattern of insulin secretion. It is concluded that [cAMP](pm) oscillations in β-cells are caused by periodic variations in cAMP generation, and that several PDEs, including PDE1, PDE3 and the IBMX-insensitive PDE8B, are required for shaping the sub-membrane cAMP signals and pulsatile insulin release.
Collapse
Affiliation(s)
- Geng Tian
- Department of Medical Cell Biology, Uppsala University, Biomedical Centre Box 571, SE-751 23 Uppsala, Sweden
| | | | | | | | | | | |
Collapse
|
114
|
Gosmain Y, Cheyssac C, Masson MH, Guérardel A, Poisson C, Philippe J. Pax6 is a key component of regulated glucagon secretion. Endocrinology 2012; 153:4204-15. [PMID: 22778220 DOI: 10.1210/en.2012-1425] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The Pax6 transcription factor is crucial for pancreatic α-cells. Indeed, Pax6-deficient mouse models are characterized by markedly altered α-cell differentiation. Our objective was to investigate the role of Pax6 in glucagon secretion process. We used a Pax6-deficient model in rat primary enriched-α cells with specific small interfering RNA leading to a 70% knockdown of Pax6 expression. We first showed that Pax6 knockdown decreases glucagon biosynthesis as well as glucagon release. Through physiological assays, we demonstrated that the decrease of Pax6 affects specifically acute glucagon secretion in primary α-cell in response to glucose, palmitate, and glucose-dependent insulinotropic peptide (GIP) but not the response to arginine and epinephrine. We identified in Pax6 knockdown model that genes involved in glucagon secretion such as the glucokinase (GCK), G protein-coupled receptor (GPR40), and GIP receptor (GIPR) as well as the corresponding proteins were significantly decreased whereas the insulin receptor (IR) Kir6.2/Sur1, and glucose transporter 1 genes were not affected. We demonstrated that Pax6 directly binds and activates specific elements on the promoter region of the GPR40, GCK, and GIPR genes. Finally, through site-directed mutagenesis experiments, we showed that disruption of Pax6 binding on the GCK, GPR40, and GIPR gene promoters led to specific decreases of their activities in the αTC1.9 glucagon-producing cell line. Hence our results indicate that Pax6 acts on the regulation of glucagon secretion at least through the transcriptional control of GCK, GPR40, and GIPR. We propose that Pax6 is not only critical for glucagon biosynthesis but also for glucagon secretion particularly in response to nutrients.
Collapse
MESH Headings
- ATP-Binding Cassette Transporters/genetics
- ATP-Binding Cassette Transporters/metabolism
- Animals
- Cells, Cultured
- Eye Proteins/genetics
- Eye Proteins/metabolism
- Glucagon/metabolism
- Glucokinase/genetics
- Glucokinase/metabolism
- Glucose Transporter Type 1/genetics
- Glucose Transporter Type 1/metabolism
- Homeodomain Proteins/genetics
- Homeodomain Proteins/metabolism
- Immunoprecipitation
- Mutagenesis, Site-Directed
- PAX6 Transcription Factor
- Paired Box Transcription Factors/genetics
- Paired Box Transcription Factors/metabolism
- Potassium Channels, Inwardly Rectifying/genetics
- Potassium Channels, Inwardly Rectifying/metabolism
- Promoter Regions, Genetic/genetics
- Protein Binding
- Rats
- Receptor, Insulin/genetics
- Receptor, Insulin/metabolism
- Receptors, Drug/genetics
- Receptors, Drug/metabolism
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
- Receptors, Gastrointestinal Hormone/genetics
- Receptors, Gastrointestinal Hormone/metabolism
- Repressor Proteins/genetics
- Repressor Proteins/metabolism
- Sulfonylurea Receptors
Collapse
Affiliation(s)
- Yvan Gosmain
- Diabetes Unit, University Hospital, University of Geneva Medical School, 1211 Geneva 14, Switzerland.
| | | | | | | | | | | |
Collapse
|
115
|
Hauge-Evans AC, Anderson RL, Persaud SJ, Jones PM. Delta cell secretory responses to insulin secretagogues are not mediated indirectly by insulin. Diabetologia 2012; 55:1995-2004. [PMID: 22526610 DOI: 10.1007/s00125-012-2546-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2012] [Accepted: 03/09/2012] [Indexed: 10/28/2022]
Abstract
AIMS/HYPOTHESIS Somatostatin from islet delta cells inhibits insulin and glucagon secretion, but knowledge of the regulation of pancreatic somatostatin release is limited. Some insulin secretagogues stimulate somatostatin secretion, and here we investigated whether delta cell secretory responses are indirectly regulated in a paracrine manner by insulin released from beta cells. METHODS Hormone release from static incubations of primary mouse islets or somatostatin-secreting TGP52 cells was measured by RIA. mRNA expression was assessed by RT-PCR. RESULTS Glucose and a range of other physiological and pharmacological agents stimulated insulin and somatostatin release, and insulin receptor mRNA was expressed in islets, MIN6 beta cells and TGP52 cells. However, exogenous insulin did not modulate basal or glucose-induced somatostatin secretion from islets, nor did pre-incubation with an antibody against the insulin receptor or with the insulin receptor tyrosine kinase inhibitor, HNMPA(AM)(3). Glucose and tolbutamide stimulated somatostatin release from TGP52 cells, whereas a range of receptor-operating agents had no effect, the latter being consistent with a lack of corresponding receptor mRNA expression in these cells. Parasympathetic activation stimulated insulin, but inhibited somatostatin release from mouse islets in accordance with differences in muscarinic receptor mRNA expression in islets, MIN6 and TGP52 cells. The inhibitory effect on somatostatin secretion was reversed by pertussis toxin or the muscarinic receptor 2 antagonist, methoctramine. CONCLUSIONS/INTERPRETATIONS A number of insulin secretagogues have analogous effects on insulin and somatostatin release, but this similarity of response is not mediated by an indirect, paracrine action of insulin on delta cells.
Collapse
Affiliation(s)
- A C Hauge-Evans
- Diabetes Research Group, Division of Diabetes and Nutritional Sciences, School of Medicine, King's College London, Guy's Campus, London Bridge, London SE1 1UL, UK.
| | | | | | | |
Collapse
|
116
|
Morris C, Banks DJ, Gaweda L, Scott S, Zhu XX, Panico M, Georgiou P, Toumazou C. A robust microfluidic in vitro cell perifusion system. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2012; 2011:8412-5. [PMID: 22256299 DOI: 10.1109/iembs.2011.6092075] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
We present here a robust microfluidic cell perifusion device for in vitro primary tissue cell secretion studies. This system increases the sample concentration to perifusion volume ratio by an order of magnitude compared with standard multi-well plate static incubation assays. Further, this device achieves physiologically relevant flow rates, pressures, and temperature. It has been manufactured with typical machining facilities, principally drilling and milling. No specialist clean room equipment is required to replicate it. We show its capability here with hormone perifusion experiments on primary pancreatic tissue from mice. This device can increase cell secretion concentrations by up to a factor of 20, allowing for the first time the direct measurement of islet glucagon using mass spectrometry.
Collapse
Affiliation(s)
- Christina Morris
- The Centre for Biologically-Inspired Technology, Bessemer Building, Level 4, Imperial College London, South Kensington Campus, London SW7 2AZ.
| | | | | | | | | | | | | | | |
Collapse
|
117
|
González-Vélez V, Dupont G, Gil A, González A, Quesada I. Model for glucagon secretion by pancreatic α-cells. PLoS One 2012; 7:e32282. [PMID: 22412861 PMCID: PMC3296707 DOI: 10.1371/journal.pone.0032282] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2011] [Accepted: 01/24/2012] [Indexed: 11/18/2022] Open
Abstract
Glucagon hormone is synthesized and released by pancreatic α-cells, one of the islet-cell types. This hormone, along with insulin, maintains blood glucose levels within the physiological range. Glucose stimulates glucagon release at low concentrations (hypoglycemia). However, the mechanisms involved in this secretion are still not completely clear. Here, using experimental calcium time series obtained in mouse pancreatic islets at low and high glucose conditions, we propose a glucagon secretion model for α-cells. Our model takes into account that the resupply of releasable granules is not only controlled by cytoplasmic , as in other neuroendocrine and endocrine cells, but also by the level of extracellular glucose. We found that, although calcium oscillations are highly variable, the average secretion rates predicted by the model fall into the range of values reported in the literature, for both stimulated and non-stimulated conditions. For low glucose levels, the model predicts that there would be a well-controlled number of releasable granules refilled slowly from a large reserve pool, probably to ensure a secretion rate that could last for several minutes. Studying the α-cell response to the addition of insulin at low glucose, we observe that the presence of insulin reduces glucagon release by decreasing the islet level. This observation is in line with previous work reporting that dynamics, mainly frequency, is altered by insulin [1]. Thus, the present results emphasize the main role played by and glucose in the control of glucagon secretion by α-cells. Our modeling approach also shows that calcium oscillations potentiate glucagon secretion as compared to constant levels of this cellular messenger. Altogether, the model sheds new light on the subcellular mechanisms involved in α-cell exocytosis, and provides a quantitative predictive tool for studying glucagon secretion modulators in physiological and pathological conditions.
Collapse
Affiliation(s)
- Virginia González-Vélez
- Departmento Ciencias Básicas, Universidad Autónoma Metropolitana Azcapotzalco, México City, México
| | - Geneviève Dupont
- Unité de Chronobiologie Théorique, Université Libre de Bruxelles, Brussels, Belgium
| | - Amparo Gil
- Departamento Matemática Aplicada y Ciencias de la Computación, Universidad de Cantabria, Santander, Cantabria, Spain
- * E-mail:
| | - Alejandro González
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Bioingeniería, Universidad Miguel Hernández, Elche, Spain
| | - Iván Quesada
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Bioingeniería, Universidad Miguel Hernández, Elche, Spain
| |
Collapse
|
118
|
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
|
119
|
Tian G, Tepikin AV, Tengholm A, Gylfe E. cAMP induces stromal interaction molecule 1 (STIM1) puncta but neither Orai1 protein clustering nor store-operated Ca2+ entry (SOCE) in islet cells. J Biol Chem 2012; 287:9862-9872. [PMID: 22298778 DOI: 10.1074/jbc.m111.292854] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The events leading to the activation of store-operated Ca(2+) entry (SOCE) involve Ca(2+) depletion of the endoplasmic reticulum (ER) resulting in translocation of the transmembrane Ca(2+) sensor protein, stromal interaction molecule 1 (STIM1), to the junctions between ER and the plasma membrane where it binds to the Ca(2+) channel protein Orai1 to activate Ca(2+) influx. Using confocal and total internal reflection fluorescence microscopy, we studied redistribution kinetics of fluorescence-tagged STIM1 and Orai1 as well as SOCE in insulin-releasing β-cells and glucagon-secreting α-cells within intact mouse and human pancreatic islets. ER Ca(2+) depletion triggered accumulation of STIM1 puncta in the subplasmalemmal ER where they co-clustered with Orai1 in the plasma membrane and activated SOCE. Glucose, which promotes Ca(2+) store filling and inhibits SOCE, stimulated retranslocation of STIM1 to the bulk ER. This effect was evident at much lower glucose concentrations in α- than in β-cells consistent with involvement of SOCE in the regulation of glucagon secretion. Epinephrine stimulated subplasmalemmal translocation of STIM1 in α-cells and retranslocation in β-cells involving raising and lowering of cAMP, respectively. The cAMP effect was mediated both by protein kinase A and exchange protein directly activated by cAMP. However, the cAMP-induced STIM1 puncta did not co-cluster with Orai1, and there was no activation of SOCE. STIM1 translocation can consequently occur independently of Orai1 clustering and SOCE.
Collapse
Affiliation(s)
- Geng Tian
- Department of Medical Cell Biology, Uppsala University, BMC Box 571, SE-751 23 Uppsala, Sweden and
| | - Alexei V Tepikin
- Physiological Laboratory, Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool L69 3BX, United Kingdom
| | - Anders Tengholm
- Department of Medical Cell Biology, Uppsala University, BMC Box 571, SE-751 23 Uppsala, Sweden and
| | - Erik Gylfe
- Department of Medical Cell Biology, Uppsala University, BMC Box 571, SE-751 23 Uppsala, Sweden and.
| |
Collapse
|
120
|
Shen XX, Li HL, Pan L, Hong J, Xiao J, Hermansen K, Jeppesen PB, Li GW. Glucotoxicity and α cell dysfunction: involvement of the PI3K/Akt pathway in glucose-induced insulin resistance in rat islets and clonal αTC1-6 cells. Endocr Res 2012; 37:12-24. [PMID: 22007944 DOI: 10.3109/07435800.2011.610855] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
AIM/HYPOTHESIS The objective of this study was to assess how long-term exposure to high glucose affects the α cell function and whether the increased glucagon secretion is mediated via insulin resistance. MATERIALS AND METHODS We established a β cell-depleted rat model to obtain pure primary α cells. Furthermore, isolated rat islets and TC1-6 cells (a clonal α cell line) were exposed to high glucose (25 or 30 mmol/L) and low glucose (5.5 mmol/L) for up to 5 days to evaluate the influence of chronic glucose toxicity on glucagon secretion and glucagon gene expression. Moreover, we added insulin and/or Wortmannin to examine if the inhibitory effect of insulin on glucagon secretion was impaired by high glucose via the phosphatidylinositol 3 kinase/PKB protein kinase B pathway. RESULTS Both glucagon secretion and glucagon gene expression were increased in response to 5 days exposure to high glucose. While a moderate insulin concentration slightly inhibits glucagon secretion from rat islets and α TC1-6 cells at high glucose, a pronounced increase in glucagon secretion was observed at low glucose. We found that the insulin-mediated activity of the phosphatidylinositol 3 kinase/PKB protein kinase B pathway in the α cell was markedly impaired by chronic exposure to high glucose. CONCLUSION The hypersecretion of glucagon induced by glucotoxicity may be secondary to insulin resistance of the α cell induced by impaired activity of the insulin signaling pathway.
Collapse
Affiliation(s)
- Xiao-Xia Shen
- Endocrinology and Cardiac Disease Clinical Center, Fuwai Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, PR China.
| | | | | | | | | | | | | | | |
Collapse
|
121
|
Yue JT, Burdett E, Coy DH, Giacca A, Efendic S, Vranic M. Somatostatin receptor type 2 antagonism improves glucagon and corticosterone counterregulatory responses to hypoglycemia in streptozotocin-induced diabetic rats. Diabetes 2012; 61:197-207. [PMID: 22106159 PMCID: PMC3237655 DOI: 10.2337/db11-0690] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Diminished responsiveness to hypoglycemia contributes to defective counterregulation in diabetes. Pancreatic and/or circulating somatostatin are elevated in diabetes, which may inhibit counterregulatory hormone release during hypoglycemia. Thus, a selective somatostatin receptor type 2 antagonist (SSTR2a) should improve hormone counterregulation to hypoglycemia. Nondiabetic (N) and streptozotocin-induced diabetic (D) rats underwent 4-h infusion of saline or SSTR2a with insulin-induced hypoglycemia clamped at 2.5 ± 0.5 mmol/L. To evaluate the effect of the SSTR2a in the absence of hypoglycemia, rats underwent a 4-h infusion of saline (Ctrl:N, Ctrl:D) or SSTR2a (Ctrl:D+SSTR2a) only. The attenuated glucagon response to hypoglycemia in D (P < 0.0002) was fully restored by SSTR2a (P < 0.0001). Furthermore, the attenuated corticosterone response in D (P < 0.002) was also enhanced by SSTR2a (P < 0.05). In the absence of hypoglycemia, SSTR2a did not alter basal blood glucose levels. D exhibited 62% more pancreatic somatostatin than N after hypoglycemia. In N rats, SSTR2a did not augment the glucagon or corticosterone response to hypoglycemia. Thus, somatostatin may contribute to impaired glucagon responsiveness to hypoglycemia in diabetes. We demonstrate that SSTR2 antagonism enhances hypoglycemia-stimulated glucagon and corticosterone release in D but not in N rats. SSTR2 antagonism does not affect basal glycemia in D rats.
Collapse
Affiliation(s)
- Jessica T.Y. Yue
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Elena Burdett
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - David H. Coy
- Department of Medicine, Peptide Research Laboratories, Tulane University, New Orleans, Louisiana
| | - Adria Giacca
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Suad Efendic
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
| | - Mladen Vranic
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Corresponding author: Mladen Vranic,
| |
Collapse
|
122
|
Hellman B, Salehi A, Grapengiesser E, Gylfe E. Isolated mouse islets respond to glucose with an initial peak of glucagon release followed by pulses of insulin and somatostatin in antisynchrony with glucagon. Biochem Biophys Res Commun 2011; 417:1219-23. [PMID: 22227186 DOI: 10.1016/j.bbrc.2011.12.113] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Accepted: 12/22/2011] [Indexed: 01/08/2023]
Abstract
Recent studies of isolated human islets have shown that glucose induces hormone release with repetitive pulses of insulin and somatostatin in antisynchrony with those of glucagon. Since the mouse is the most important animal model we studied the temporal relation between hormones released from mouse islets. Batches of 5-10 islets were perifused and the hormones measured with radioimmunoassay in 30s fractions. At 3mM glucose, hormone secretion was stable with no detectable pulses of glucagon, insulin or somatostatin. Increase of glucose to 20mM resulted in an early secretory phase with a glucagon peak followed by peaks of insulin and somatostatin. Subsequent hormone secretion was pulsatile with a periodicity of 5min. Cross-correlation analyses showed that the glucagon pulses were antisynchronous to those of insulin and somatostatin. In contrast to the marked stimulation of insulin and somatostatin secretion, the pulsatility resulted in inhibition of overall glucagon release. The cytoarchitecture of mouse islets differs from that of human islets, which may affect the interactions between the hormone-producing cells. Although indicating that paracrine regulation is important for the characteristic patterns of pulsatile hormone secretion, the mouse data mimic those of human islets with more than 20-fold variations of the insulin/glucagon ratio. The data indicate that the mouse serves as an appropriate animal model for studying the temporal relation between the islet hormones controlling glucose production in the liver.
Collapse
Affiliation(s)
- Bo Hellman
- Department of Medical Cell Biology, University of Uppsala, SE-75123 Uppsala, Sweden.
| | | | | | | |
Collapse
|
123
|
Yashiro H, Tsujihata Y, Takeuchi K, Hazama M, Johnson PRV, Rorsman P. The effects of TAK-875, a selective G protein-coupled receptor 40/free fatty acid 1 agonist, on insulin and glucagon secretion in isolated rat and human islets. J Pharmacol Exp Ther 2011; 340:483-9. [PMID: 22106100 DOI: 10.1124/jpet.111.187708] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
G protein-coupled receptor 40 (GPR40)/free fatty acid 1 (FFA1) is a G protein-coupled receptor involved in free fatty acid-induced insulin secretion. To analyze the effect of our novel GPR40/FFA1-selective agonist, [(3S)-6-({2',6'-dimethyl-4'-[3-(methylsulfonyl)propoxy]biphenyl-3-yl}methoxy)-2,3-dihydro-1-benzofuran-3-yl]acetic acid hemi-hydrate (TAK-875), on insulin and glucagon secretion, we performed hormone secretion assays and measured intracellular Ca²⁺ concentration ([Ca²⁺](i)) in both human and rat islets. Insulin and glucagon secretion were measured in static and dynamic conditions by using groups of isolated rat and human pancreatic islets. [Ca²⁺](i) was recorded by using confocal microscopy. GPR40/FFA1 expression was measured by quantitative polymerase chain reaction. In both human and rat islets, TAK-875 enhanced glucose-induced insulin secretion in a glucose-dependent manner. The stimulatory effect of TAK-875 was similar to that produced by glucagon-like peptide-1 and correlated with the elevation of β-cell [Ca²⁺](i). TAK-875 was without effect on glucagon secretion at both 1 and 16 mM glucose in human islets. These data indicate that GPR40/FFA1 influences mainly insulin secretion in a glucose-dependent manner. The β-cell-specific action of TAK-875 in human islets may represent a therapeutically useful feature that allows plasma glucose control without compromising counter-regulation of glucagon secretion, thus minimizing the risk of hypoglycemia.
Collapse
Affiliation(s)
- Hiroaki Yashiro
- Metabolic Disease Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 17-85 Jusohonmachi 2-chome, Yodogawa-ku, Osaka 532-8686, Japan.
| | | | | | | | | | | |
Collapse
|
124
|
Bagger JI, Knop FK, Holst JJ, Vilsbøll T. Glucagon antagonism as a potential therapeutic target in type 2 diabetes. Diabetes Obes Metab 2011; 13:965-71. [PMID: 21615669 DOI: 10.1111/j.1463-1326.2011.01427.x] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Glucagon is a hormone secreted from the alpha cells of the pancreatic islets. Through its effect on hepatic glucose production (HGP), glucagon plays a central role in the regulation of glucose homeostasis. In patients with type 2 diabetes mellitus (T2DM), abnormal regulation of glucagon secretion has been implicated in the development of fasting and postprandial hyperglycaemia. Therefore, new therapeutic agents based on antagonizing glucagon action, and hence blockade of glucagon-induced HGP, could be effective in lowering both fasting and postprandial hyperglycaemia in patients with T2DM. This review focuses on the mechanism of action, safety and efficacy of glucagon antagonists in the treatment of T2DM and discusses the challenges associated with this new potential antidiabetic treatment modality.
Collapse
Affiliation(s)
- J I Bagger
- Diabetes Research Division, Department of Internal Medicine F, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
| | | | | | | |
Collapse
|
125
|
Henquin JC, Accili D, Ahrén B, Boitard C, Seino S, Cerasi E. Long in the shade, glucagon re-occupies centre court. Diabetes Obes Metab 2011; 13 Suppl 1:v-viii. [PMID: 21824249 DOI: 10.1111/j.1463-1326.2011.01476.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
126
|
Kawamori D, Akiyama M, Hu J, Hambro B, Kulkarni RN. Growth factor signalling in the regulation of α-cell fate. Diabetes Obes Metab 2011; 13 Suppl 1:21-30. [PMID: 21824253 DOI: 10.1111/j.1463-1326.2011.01442.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Glucagon plays critical roles in regulating glucose homeostasis, mainly by counteracting the effects of insulin. Consequently, the dysregulated glucagon secretion that is evident in type 2 diabetes has significant implications in the pathophysiology of the disease. Glucagon secretion from pancreatic α-cells has been suggested to be modulated by blood glucose, signals from the nervous system and endocrine components. In addition to these regulators, intraislet factors acting in a paracrine manner from neighbouring β-cells are emerging as central modulator(s) of α-cell biology. One of the most important of these paracrine factors, insulin, modulates glucagon secretion. Indeed, the α-cell-specific insulin receptor knockout (αIRKO) mouse manifests hypersecretion of glucagon in the postprandial stage and exhibits defective secretion in fasting-induced hypoglycaemia, together mimicking the α-cell defects observed in type 2 diabetes. Interestingly, αIRKO mice display a progressive increase in β-cell mass and a concomitant decrease in α-cells. Lineage trace analyses reveal that the new β-cells originate, in part, from the insulin receptor-deficient α-cells indicating a critical role for α-cell insulin signalling in determining β-cell origin. Our studies also reveal that glucagon-like peptide-1 (GLP-1) treatment of αIRKO mice suppresses glucagon secretion despite absence of functional insulin receptors precluding a role for insulin in GLP-1 action on α-cells in this model. These findings highlight the significance of insulin signalling in the regulation of α-cell biology.
Collapse
Affiliation(s)
- D Kawamori
- Section of Islet Cell Biology and Regenerative Medicine, Joslin Diabetes Center and Harvard Medical School, Boston, MA 02215, USA.
| | | | | | | | | |
Collapse
|
127
|
Hardy AB, Serino AS, Wijesekara N, Chimienti F, Wheeler MB. Regulation of glucagon secretion by zinc: lessons from the β cell-specific Znt8 knockout mouse model. Diabetes Obes Metab 2011; 13 Suppl 1:112-7. [PMID: 21824264 DOI: 10.1111/j.1463-1326.2011.01451.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
In type-2 diabetes, hyperglucagonaemia aggravates elevated blood glucose levels. Relative to our knowledge of the β-cell and insulin secretion, there remains a limited understanding of glucagon secretion in α-cells. Regulation of glucagon may be dependent on a combination of factors, which include direct glucose sensing by the α-cell, innervations from the autonomic nervous system and potential 'paracrine' actions by hormones and factors that are released by adjacent endocrine cells within the islets. The list of potential 'paracrine' regulators within the islet includes insulin, somatostatin, γ-aminobutyric acid, glutamate and zinc. Zinc crystallises with insulin in β-cells and is co-secreted with insulin. In the scientific literature, the effect of exogeneous zinc on glucagon secretion has been debated. Here, we confirm that an increase in exogeneous zinc does inhibit glucagon secretion. To determine if there are physiological effects of zinc on glucagon secretion we used a β-cell-specific ZnT8 knockout (Znt8BKO) mouse model. Znt8BKO mice, despite showing lower granular zinc content in β-cells, showed no changes in fasted plasma glucagon levels and glucose regulated glucagon secretion. These findings suggest that zinc secreted from β-cell does not regulate glucagon secretion.
Collapse
Affiliation(s)
- A B Hardy
- Department of Physiology, University of Toronto, 1 King's College Circle, Toronto, ON, Canada
| | | | | | | | | |
Collapse
|
128
|
Hetherington J, Sumner T, Seymour RM, Li L, Rey MV, Yamaji S, Saffrey P, Margoninski O, Bogle IDL, Finkelstein A, Warner A. A composite computational model of liver glucose homeostasis. I. Building the composite model. J R Soc Interface 2011; 9:689-700. [PMID: 21676967 DOI: 10.1098/rsif.2011.0141] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
A computational model of the glucagon/insulin-driven liver glucohomeostasis function, focusing on the buffering of glucose into glycogen, has been developed. The model exemplifies an 'engineering' approach to modelling in systems biology, and was produced by linking together seven component models of separate aspects of the physiology. The component models use a variety of modelling paradigms and degrees of simplification. Model parameters were determined by an iterative hybrid of fitting to high-scale physiological data, and determination from small-scale in vitro experiments or molecular biological techniques. The component models were not originally designed for inclusion within such a composite model, but were integrated, with modification, using our published modelling software and computational frameworks. This approach facilitates the development of large and complex composite models, although, inevitably, some compromises must be made when composing the individual models. Composite models of this form have not previously been demonstrated.
Collapse
Affiliation(s)
- J Hetherington
- CoMPLEX, University College London, Gower Street, London WC1E 6BT, UK
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
129
|
Lund A, Vilsbøll T, Bagger JI, Holst JJ, Knop FK. The separate and combined impact of the intestinal hormones, GIP, GLP-1, and GLP-2, on glucagon secretion in type 2 diabetes. Am J Physiol Endocrinol Metab 2011; 300:E1038-46. [PMID: 21386059 DOI: 10.1152/ajpendo.00665.2010] [Citation(s) in RCA: 123] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Type 2 diabetes mellitus (T2DM) is associated with reduced suppression of glucagon during oral glucose tolerance test (OGTT), whereas isoglycemic intravenous glucose infusion (IIGI) results in normal glucagon suppression in these patients. We examined the role of the intestinal hormones glucose-dependent insulinotropic polypeptide (GIP), glucagon-like peptide-1 (GLP-1), and glucagon-like peptide-2 (GLP-2) in this discrepancy. Glucagon responses were measured during a 3-h 50-g OGTT (day A) and an IIGI (day B) in 10 patients with T2DM [age (mean ± SE), 51 ± 3 yr; body mass index, 33 ± 2 kg/m(2); HbA(1c), 6.5 ± 0.2%]. During four additional IIGIs, GIP (day C), GLP-1 (day D), GLP-2 (day E) and a combination of the three (day F) were infused intravenously. Isoglycemia during all six study days was obtained. As expected, no suppression of glucagon occurred during the initial phase of the OGTT, whereas significantly (P < 0.05) lower plasma levels of glucagon during the first 30 min of the IIGI (day B) were observed. The glucagon response during the IIGI + GIP + GLP-1 + GLP-2 infusion (day F) equaled the inappropriate glucagon response to OGTT (P = not significant). The separate GIP infusion (day C) elicited significant hypersecretion of glucagon, whereas GLP-1 infusion (day D) resulted in enhancement of glucagon suppression during IIGI. IIGI + GLP-2 infusion (day E) resulted in a glucagon response in the midrange between the glucagon responses to OGTT and IIGI. Our results indicate that the intestinal hormones, GIP, GLP-1, and GLP-2, may play a role in the inappropriate glucagon response to orally ingested glucose in T2DM with, especially, GIP, acting to increase glucagon secretion.
Collapse
Affiliation(s)
- Asger Lund
- Diabetes Research Division, Department of Internal Medicine F, Gentofte Hospital, Hellerup, Denmark
| | | | | | | | | |
Collapse
|
130
|
Gao S, Oh YB, Shah A, Park WH, Kim SH. Suppression of ANP secretion by somatostatin through somatostatin receptor type 2. Peptides 2011; 32:1179-86. [PMID: 21539874 DOI: 10.1016/j.peptides.2011.04.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2011] [Revised: 04/15/2011] [Accepted: 04/15/2011] [Indexed: 12/11/2022]
Abstract
Somatostatin is a cyclic-14 amino acid peptide which mainly distributed in digestive system and brain. Somatostatin receptor (SSTR) is a G-protein coupled receptor and all five SSTR subtypes are expressed in cardiomyocytes. The aim of this study was to investigate the effect of somatostatin on atrial natriuretic peptide (ANP) secretion and its signaling pathway. Somatostatin (0.01 and 0.1nM) decreased ANP secretion in isolated beating rat atrium in a dose-dependent manner. But atrial contractility and translocation of extracellular fluid were not changed. Somatostatin-induced decrease in ANP secretion was significantly attenuated by the pretreatment with CYN 154806 (SSTR type 2 antagonist; 0.1μM), but not by BIM 23056 (SSTR type 5 antagonist; 0.1μM) and urantide (urotensin II receptor antagonist; 0.1μM). When pretreated with an agonist for SSTR type 2 (Seglitide, 0.1nM) and SSTR type 5 (L 817818, 0.1nM), only Seglitide reduced ANP secretion similar to that of somatostatin. The suppressive effect of somatostatin on ANP secretion was attenuated by the pretreatment with an inhibitor for adenylyl cyclase (MDL-12330A, 5μM) or protein kinase A (KT 5720, 0.1μM). In diabetic rat atria, the suppressive effect of somatostatin on ANP secretion and concentration was attenuated. Real time-PCR and western blot shows the decreased level of SSTR type 2 mRNA and protein in diabetic rat atria. These data suggest that somatostatin decreased ANP secretion through SSTR type 2 and an attenuation of suppressive effect of somatostatin on ANP secretion in diabetic rat atria is due to a down-regulation of SSTR type 2.
Collapse
Affiliation(s)
- Shan Gao
- Department of Physiology, Research Institute for Endocrine Sciences, Chonbuk National University Medical School, 2-20 Keum-Am-Dong-San, Jeonju 561-180, Republic of Korea
| | | | | | | | | |
Collapse
|
131
|
Tian G, Sandler S, Gylfe E, Tengholm A. Glucose- and hormone-induced cAMP oscillations in α- and β-cells within intact pancreatic islets. Diabetes 2011; 60:1535-43. [PMID: 21444924 PMCID: PMC3292328 DOI: 10.2337/db10-1087] [Citation(s) in RCA: 114] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE cAMP is a critical messenger for insulin and glucagon secretion from pancreatic β- and α-cells, respectively. Dispersed β-cells show cAMP oscillations, but the signaling kinetics in cells within intact islets of Langerhans is unknown. RESEARCH DESIGN AND METHODS The subplasma-membrane cAMP concentration ([cAMP](pm)) was recorded in α- and β-cells in the mantle of intact mouse pancreatic islets using total internal reflection microscopy and a fluorescent translocation biosensor. Cell identification was based on the opposite effects of adrenaline on cAMP in α- and β-cells. RESULTS In islets exposed to 3 mmol/L glucose, [cAMP](pm) was low and stable. Glucagon and glucagon-like peptide-1(7-36)-amide (GLP-1) induced dose-dependent elevation of [cAMP](pm), often with oscillations synchronized among β-cells. Whereas glucagon also induced [cAMP](pm) oscillations in most α-cells, <20% of the α-cells responded to GLP-1. Elevation of the glucose concentration to 11-30 mmol/L in the absence of hormones induced slow [cAMP](pm) oscillations in both α- and β-cells. These cAMP oscillations were coordinated with those of the cytoplasmic Ca(2+) concentration ([Ca(2+)](i)) in the β-cells but not caused by the changes in [Ca(2+)](i). The transmembrane adenylyl cyclase (AC) inhibitor 2'5'-dideoxyadenosine suppressed the glucose- and hormone-induced [cAMP](pm) elevations, whereas the preferential inhibitors of soluble AC, KH7, and 1,3,5(10)-estratrien-2,3,17-β-triol perturbed cell metabolism and lacked effect, respectively. CONCLUSIONS Oscillatory [cAMP](pm) signaling in secretagogue-stimulated β-cells is maintained within intact islets and depends on transmembrane AC activity. The discovery of glucose- and glucagon-induced [cAMP](pm) oscillations in α-cells indicates the involvement of cAMP in the regulation of pulsatile glucagon secretion.
Collapse
|
132
|
Abstract
OBJECTIVE The physiologic significance of the nitric oxide (NO)/cGMP signaling pathway in islets is unclear. We hypothesized that cGMP-dependent protein kinase type I (cGKI) is directly involved in the secretion of islet hormones and glucose homeostasis. RESEARCH DESIGN AND METHODS Gene-targeted mice that lack cGKI in islets (conventional cGKI mutants and cGKIα and Iβ rescue mice [α/βRM] that express cGKI only in smooth muscle) were studied in comparison to control (CTR) mice. cGKI expression was mapped in the endocrine pancreas by Western blot, immuno-histochemistry, and islet-specific recombination analysis. Insulin, glucagon secretion, and cytosolic Ca²(+) ([Ca²(+)](i)) were assayed by radioimmunoassay and FURA-2 measurements, respectively. Serum levels of islet hormones were analyzed at fasting and upon glucose challenge (2 g/kg) in vivo. RESULTS Immunohistochemistry showed that cGKI is present in α- but not in β-cells in islets of Langerhans. Mice that lack α-cell cGKI had significantly elevated fasting glucose and glucagon levels, whereas serum insulin levels were unchanged. High glucose concentrations strongly suppressed the glucagon release in CTR mice, but had only a moderate effect on islets that lacked cGKI. 8-Br-cGMP reduced stimulated [Ca²(+)](i) levels and glucagon release rates of CTR islets at 0.5 mmol/l glucose, but was without effect on [Ca²(+)](i) or hormone release in cGKI-deficient islets. CONCLUSIONS We propose that cGKI modulates glucagon release by suppression of [Ca²(+)](i) in α-cells.
Collapse
Affiliation(s)
- Veronika Leiss
- FOR 923, Technische Universität München, München, Germany, and Center for Integrated Protein Science, Ludwig-Maximilians-Universität München, München, Germany
- Institut für Pharmakologie und Toxikologie, Abteilung Pharmakologie und Experimentelle Therapie, Universitätsklinikum Tübingen, Tübingen, Germany
| | - Andreas Friebe
- Lehrstuhl für Physiologie I, Julius-Maximilians Universität Würzburg, Würzburg, Germany
| | - Andrea Welling
- FOR 923, Technische Universität München, München, Germany, and Center for Integrated Protein Science, Ludwig-Maximilians-Universität München, München, Germany
- Institut für Pharmakologie und Toxikologie, Technische Universität München, München, Germany
| | - Franz Hofmann
- FOR 923, Technische Universität München, München, Germany, and Center for Integrated Protein Science, Ludwig-Maximilians-Universität München, München, Germany
| | - Robert Lukowski
- FOR 923, Technische Universität München, München, Germany, and Center for Integrated Protein Science, Ludwig-Maximilians-Universität München, München, Germany
- Institut für Pharmazie, Abteilung Pharmakologie, Toxikologie und Klinische Pharmazie, Universität Tübingen, Tübingen, Germany
- Corresponding author: Robert Lukowski,
| |
Collapse
|
133
|
Hauge-Evans AC, King AJ, Fairhall K, Persaud SJ, Jones PM. A role for islet somatostatin in mediating sympathetic regulation of glucagon secretion. Islets 2010; 2:341-4. [PMID: 21099335 PMCID: PMC3062248 DOI: 10.4161/isl.2.6.13858] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
AIMS/HYPOTHESIS Somatostatin (SST) released from islet δ-cells inhibits both insulin and glucagon secretion but the role of this tonic inhibition is unclear. In this study we investigated whether δ-cell SST may facilitate sympathetic regulation of glucagon secretion as part of an 'accelerator/brake' mechanism. METHODS The secretory characteristics of islets isolated from SST-deficient (Sst-/-) and control mouse islets were assessed in static incubation studies. Glucagon and SST release was measured by radioimmunoassay (RIA). RESULTS Arginine stimulated both glucagon and SST release from control mouse islets whereas the sympathetic neurotransmitter noradrenaline (NA) increased glucagon secretion but inhibited SST release in the presence of 2 mmol/l glucose or 20 mmol/l arginine. Experiments were performed using Sst-/- islets to assess whether the reduction of SST secretion by NA offers an indirect mechanism of enhancing glucagon release in response to sympathetic activation. Arginine-induced but not NA-induced glucagon release from Sst-/- islets was significantly increased compared to controls. In combination, NA enhanced arginine-induced release from both groups of mouse islets but to a greater extent in control islets, leading to similar overall levels of glucagon release. The responsiveness of Sst-/- islets to NA was thus blunted under stimulatory but not sub-stimulatory conditions of SST release. CONCLUSIONS Our data suggest that sympathetic activation of glucagon release may be partly mediated by an indirect effect on SST secretion, where the tonic inhibition by δ-cell SST on α-cells is removed, facilitating precise and substantial changes in glucagon release in response to NA.
Collapse
|
134
|
Kelly C, Flatt PR, McClenaghan NH. Cell-to-cell communication and cellular environment alter the somatostatin status of delta cells. Biochem Biophys Res Commun 2010; 399:162-6. [PMID: 20637727 DOI: 10.1016/j.bbrc.2010.07.036] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2010] [Accepted: 07/12/2010] [Indexed: 11/17/2022]
Abstract
INTRODUCTION Somatostatin, released from pancreatic delta cells, is a potent paracrine inhibitor of insulin and glucagon secretion. Islet cellular interactions and glucose homeostasis are essential to maintain normal patterns of insulin secretion. However, the importance of cell-to-cell communication and cellular environment in the regulation of somatostatin release remains unclear. METHODS This study employed the somatostatin-secreting TGP52 cell line maintained in DMEM:F12 (17.5mM glucose) or DMEM (25mM glucose) culture media. The effect of pseudoislet formation and culture medium on somatostatin content and release in response to a variety of stimuli was measured by somatostatin EIA. In addition, the effect of pseudoislet formation on cellular viability (MTT and LDH assays) and proliferation (BrdU ELISA) was determined. RESULTS TGP52 cells readily formed pseudoislets and showed enhanced functionality in three-dimensional form with increased E-cadherin expression irrespective of the culture environment used. However, culture in DMEM decreased cellular somatostatin content (P<0.01) and increased somatostatin secretion in response to a variety of stimuli including arginine, calcium and PMA (P<0.001) when compared with cells grown in DMEM:F12. Configuration of TGP52 cells as pseudoislets reduced the proliferative rate and increased cellular cytotoxicity irrespective of culture medium used. CONCLUSIONS Somatostatin secretion is greatly facilitated by cell-to-cell interactions and E-cadherin expression. Cellular environment and extracellular glucose also significantly influence the function of delta cells.
Collapse
Affiliation(s)
- Catriona Kelly
- SAAD Centre for Pharmacy & Diabetes, School of Biomedical Sciences, University of Ulster, Coleraine, UK.
| | | | | |
Collapse
|
135
|
Balhuizen A, Kumar R, Amisten S, Lundquist I, Salehi A. Activation of G protein-coupled receptor 30 modulates hormone secretion and counteracts cytokine-induced apoptosis in pancreatic islets of female mice. Mol Cell Endocrinol 2010; 320:16-24. [PMID: 20122988 DOI: 10.1016/j.mce.2010.01.030] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2009] [Revised: 01/21/2010] [Accepted: 01/21/2010] [Indexed: 12/22/2022]
Abstract
The role of the newly discovered estrogen receptor GPR30 in islet physiology and pathophysiology is unclear. We examined GPR30 expression in relation to hormone secretion and possible anti-apoptotic effects in isolated mouse islets using the synthetic GPR30 ligand G-1. The mRNA and protein expression of GPR30 was analyzed by qPCR, Western blot and confocal microscopy. Hormone secretion and cAMP content were determined with RIA and apoptosis in islet cells with the Annexin-V method. GPR30 mRNA and protein expression was markedly higher in islets from females compared to male. This gender difference was not found for the genomic estrogen receptors ER alpha and ER beta, the ER alpha expression being 10-fold higher than ER beta in both genders. Confocal microscopy revealed abounden GPR30 expression in insulin, glucagon and somatostatin cells. Dose-response studies of G-1 vs 17beta-estradiol in isolated islets at 1 or 12 mM glucose showed an almost identical pattern in that both compounds increased insulin and inhibited glucagon and somatostatin secretion. ICI-182,780 and EM-652, potent antagonists of the 17beta-estradiol receptors (ER alpha and ER beta) did not influence the amplifying effect of G-1 or 17beta-estradiol on cAMP content or insulin secretion from isolated islets. Cytokine-induced (IL-1 beta+TNFalpha+INF gamma) apoptosis in islets, cultured for 24h at 5mM glucose, was almost abolished by G-1 or 17beta-estradiol treatment. Addition of ICI-182,780 or EM-652 did not affect this beneficial effect of G-1 or 17beta-estradiol. Taken together, our findings show that GPR30 is expressed in most islet endocrine cells. The synthetic GPR30 ligand G-1 mimics the non-genomic effects of 17beta-estradiol on islet hormone secretion, cAMP content in islets and its anti-apoptotic effects. G-1 or analogs thereof might be new potential candidates in the therapeutic strategy for type 2 diabetes in women.
Collapse
Affiliation(s)
- Alexander Balhuizen
- Department of Clinical Science, Islet Cell Physiology, University of Lund, Sweden
| | | | | | | | | |
Collapse
|
136
|
Soria B, Tudurí E, González A, Hmadcha A, Martin F, Nadal A, Quesada I. Pancreatic islet cells: a model for calcium-dependent peptide release. HFSP JOURNAL 2010; 4:52-60. [PMID: 20885773 DOI: 10.2976/1.3364560] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2010] [Accepted: 02/25/2010] [Indexed: 11/19/2022]
Abstract
In mammals the concentration of blood glucose is kept close to 5 mmol∕l. Different cell types in the islet of Langerhans participate in the control of glucose homeostasis. β-cells, the most frequent type in pancreatic islets, are responsible for the synthesis, storage, and release of insulin. Insulin, released with increases in blood glucose promotes glucose uptake into the cells. In response to glucose changes, pancreatic α-, β-, and δ-cells regulate their electrical activity and Ca(2+) signals to release glucagon, insulin, and somatostatin, respectively. While all these signaling steps are stimulated in hypoglycemic conditions in α-cells, the activation of these events require higher glucose concentrations in β and also in δ-cells. The stimulus-secretion coupling process and intracellular Ca(2+) ([Ca(2+)](i)) dynamics that allow β-cells to secrete is well-accepted. Conversely, the mechanisms that regulate α- and δ-cell secretion are still under study. Here, we will consider the glucose-induced signaling mechanisms in each cell type and the mathematical models that explain Ca(2+) dynamics.
Collapse
|
137
|
Le Marchand SJ, Piston DW. Glucose suppression of glucagon secretion: metabolic and calcium responses from alpha-cells in intact mouse pancreatic islets. J Biol Chem 2010; 285:14389-98. [PMID: 20231269 DOI: 10.1074/jbc.m109.069195] [Citation(s) in RCA: 117] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Glucagon is released from alpha-cells present in intact pancreatic islets at glucose concentrations below 4 mm, whereas higher glucose levels inhibit its secretion. The mechanisms underlying the suppression of alpha-cell secretory activity are poorly understood, but two general types of models have been proposed as follows: direct inhibition by glucose or paracrine inhibition from non-alpha-cells within the islet of Langerhans. To identify alpha-cells for analysis, we utilized transgenic mice expressing fluorescent proteins targeted specifically to these cells. Measurements of glucagon secretion from pure populations of flow-sorted alpha-cells show that contrary to its effect on intact islets, glucose does stimulate glucagon secretion from isolated alpha-cells. This observation argues against a direct inhibition of glucagon secretion by glucose and supports the paracrine inhibition model. Imaging of cellular metabolism by two-photon excitation of NAD(P)H autofluorescence indicates that glucose is metabolized in alpha-cells and that glucokinase is the likely rate-limiting step in this process. Imaging calcium dynamics of alpha-cells in intact islets reveals that inhibiting concentrations of glucose increase the intracellular calcium concentration and the frequency of alpha-cell calcium oscillations. Application of candidate paracrine inhibitors leads to reduced glucagon secretion but did not decrease the alpha-cell calcium activity. Taken together, the data suggest that suppression occurs downstream from alpha-cell calcium signaling, presumably at the level of vesicle trafficking or exocytotic machinery.
Collapse
Affiliation(s)
- Sylvain J Le Marchand
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee 37232, USA
| | | |
Collapse
|
138
|
Kawamori D, Welters HJ, Kulkarni RN. Molecular Pathways Underlying the Pathogenesis of Pancreatic α-Cell Dysfunction. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 654:421-45. [DOI: 10.1007/978-90-481-3271-3_18] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
139
|
Slucca M, Harmon JS, Oseid EA, Bryan J, Robertson RP. ATP-sensitive K+ channel mediates the zinc switch-off signal for glucagon response during glucose deprivation. Diabetes 2010; 59:128-34. [PMID: 19808893 PMCID: PMC2797913 DOI: 10.2337/db09-1098] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE The intraislet insulin hypothesis proposes that glucagon secretion during hypoglycemia is triggered by a decrease in intraislet insulin secretion. A more recent hypothesis based on in vivo data from hypoglycemic rats is that it is the decrease in zinc cosecreted with insulin from beta-cells, rather than the decrease in insulin itself, that signals glucagon secretion from alpha-cells during hypoglycemia. These studies were designed to determine whether closure of the alpha-cell ATP-sensitive K(+) channel (K(ATP) channel) is the mechanism through which the zinc switch-off signal triggers glucagon secretion during glucose deprivation. RESEARCH DESIGN AND METHODS All studies were performed using perifused isolated islets. RESULTS In control experiments, the expected glucagon response to an endogenous insulin switch-off signal during glucose deprivation was observed in wild-type mouse islets. In experiments with streptozotocin-treated wild-type islets, a glucagon response to an exogenous zinc switch-off signal was observed during glucose deprivation. However, this glucagon response to the zinc switch-off signal during glucose deprivation was not seen in the presence of nifedipine, diazoxide, or tolbutamide or if K(ATP) channel knockout mouse islets were used. All islets had intact glucagon responses to epinephrine. CONCLUSIONS These data demonstrate that closure of K(ATP) channels and consequent opening of calcium channels is the mechanism through which the zinc switch-off signal triggers glucagon secretion during glucose deprivation.
Collapse
Affiliation(s)
- Michela Slucca
- From the Pacific Northwest Diabetes Research Institute and the Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine and Department of Pharmacology, University of Washington, Seattle, Washington
| | - Jamie S. Harmon
- From the Pacific Northwest Diabetes Research Institute and the Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine and Department of Pharmacology, University of Washington, Seattle, Washington
| | - Elizabeth A. Oseid
- From the Pacific Northwest Diabetes Research Institute and the Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine and Department of Pharmacology, University of Washington, Seattle, Washington
| | - Joseph Bryan
- From the Pacific Northwest Diabetes Research Institute and the Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine and Department of Pharmacology, University of Washington, Seattle, Washington
| | - R. Paul Robertson
- From the Pacific Northwest Diabetes Research Institute and the Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine and Department of Pharmacology, University of Washington, Seattle, Washington
- Corresponding author: R. Paul Robertson,
| |
Collapse
|
140
|
Hellman B, Salehi A, Gylfe E, Dansk H, Grapengiesser E. Glucose generates coincident insulin and somatostatin pulses and antisynchronous glucagon pulses from human pancreatic islets. Endocrinology 2009; 150:5334-40. [PMID: 19819962 DOI: 10.1210/en.2009-0600] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The kinetics of insulin, glucagon and somatostatin release was studied in human pancreatic islets. Batches of 10-15 islets were perifused and the hormones measured with RIA in 30-sec fractions. Increase of glucose from 3 to 20 mm resulted in a brief pulse of glucagon coinciding with suppression of basal insulin and somatostatin release. There was a subsequent drop of glucagon release concomitant with the appearance of a pronounced pulse of insulin and a slightly delayed pulse of somatostatin. Continued exposure to 20 mm glucose generated pulsatile release of the three hormones with 7- to 8-min periods accounting for 60-70% of the secreted amounts. Glucose caused pronounced stimulation of average insulin and somatostatin release. However, the nadirs between the glucagon pulses were lower than the secretion at 3 mm glucose, resulting in 18% suppression of average release. The repetitive glucagon pulses were antisynchronous to coincident pulses of insulin and somatostatin. The resulting greater than 20-fold variations of the insulin to glucagon ratio might be essential for minute-to-minute regulation of the hepatic glucose production.
Collapse
Affiliation(s)
- Bo Hellman
- Department of Medical Cell Biology, University of Uppsala, Uppsala, Sweden.
| | | | | | | | | |
Collapse
|
141
|
Lau YY, Ma P, Gibiansky L, Komorowski R, Wang J, Wang G, Yan H, Véniant MM, Kakkar T. Pharmacokinetic and pharmacodynamic modeling of a monoclonal antibody antagonist of glucagon receptor in male ob/ob mice. AAPS JOURNAL 2009; 11:700-9. [PMID: 19851873 DOI: 10.1208/s12248-009-9150-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2009] [Accepted: 09/24/2009] [Indexed: 01/04/2023]
Abstract
Elevated basal concentrations of glucagon and reduced postprandial glucagon suppression are partly responsible for the increased hepatic glucose production seen in type 2 diabetic patients. Recently, it was demonstrated that an antagonistic human monoclonal antibody (mAb) blocking glucagon receptor (GCGR) has profound glucose-lowering effects in various animal models. To further understand the effects on glucose homeostasis mediated by such an antibody, a pharmacokinetic-pharmacodynamic (PK-PD) study was conducted in a diabetic ob/ob mouse model. Four groups of ob/ob mice were randomized to receive single intraperitoneal administration of placebo, 0.6, 1, or 3 mg/kg of mAb GCGR, a fully human mAb against GCGR. The concentration-time data were used for noncompartmental and compartmental analysis. A semi-mechanistic PK-PD model incorporating the glucose-glucagon inter-regulation and the hypothesized inhibitory effect of mAb GCGR on GCGR signaling pathway via competitive inhibition was included to describe the disposition of glucose and glucagon over time. The pharmacokinetics of mAb GCGR was well characterized by a two-compartment model with parallel linear and nonlinear saturable eliminations. Single injection of mAb GCGR caused a rapid glucose-lowering effect with blood glucose concentrations returning to baseline by 4 to 18 days with increasing dose from 0.6 to 3 mg/kg. Elevation of glucagon concentrations was also observed in a dose-dependent manner. The results illustrated that the feedback relationship between glucose and glucagon in the presence of mAb GCGR could be quantitatively described by the developed model. The model may provide additional understanding in the underlying mechanism of GCGR antagonism by mAb.
Collapse
Affiliation(s)
- Yvonne Y Lau
- Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., One Amgen Center Dr., Thousand Oaks, California 91320, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
142
|
Dufrane D, Maillart JF, Aouassar N, Goebbels RM, Guiot Y, Gianello P. Native pancreatic alpha-cell adaptation in streptozotocin-induced diabetic primates: importance for pig islet xenotransplantation. Xenotransplantation 2009; 16:152-63. [PMID: 19566655 DOI: 10.1111/j.1399-3089.2009.00530.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND Metabolic compatibility between donor and recipient species is an important matter for pig islet xenotransplantation. Glucagon is a key hormone for the function of pig islets as well as control of hypoglycemia in the recipients of the islets. Because a discrepancy exists in the composition of glucagon cells of pig and human/primate islets, the present study was designed to determine the role of native recipient glucagon cells in the treatment of diabetes by islet transplantation in a "pig-to-primate" model. METHODS Streptozotocin-treated (50 mg/kg) monkeys (n = 12, follow-up of 6 to 231 days) were compared with non-diabetic animals (n = 5; follow-up, 180 days). Metabolic [fasting and intravenous glucose tolerance tests (IVGTTs) for serum levels of glucose, insulin, glucagon] and morphologic (endocrine volume density and cell mass for insulin and glucagon) were compared between non-diabetic and diabetic animals. Six additional diabetic primates were given transplants of 15 000 adult pig islet equivalents without immunosuppression to monitor glucose, glucagon, insulin, and porcine C-peptide levels until 48 h after transplantation. RESULTS Elevated fasting blood glucose, pathologic IVGTT, destruction of 95% of beta-cell mass, and glycosylated hemoglobin (>13%) were assessed in diabetic monkeys. The serum glucagon levels and glucagon cell mass correlated significantly with diabetes time course of diabetes (R = 0.940, p = 0.005; R = 0.663, p = 0.019, respectively). A mean increase of 89% in glucagon cell mass was observed for primates suffering from diabetes >53 days. No response of glucagon secretion was observed for diabetic animals during IVGTT, because no increase of serum insulin levels followed glucose loading. Blood glucose levels dropped after pig islet xenografts in diabetic primates. This reduction was maintained by an insulin level >20 microU/ml over the period of time of xenograft function (porcine C-peptide >0.1 ng/ml). A total restoration of native primate glucagon sensitivity to insulin was found after pig islets xenotransplantation as revealed by a reduction of 80% of the glucagon level. When graft dysfunction (>24 h post-transplantation), the insulin level dropped and glucagon levels rose again (>50 pg/ml). CONCLUSIONS Native glucagon cells provide morphologic and functional plasticity to diabetes. Adult pig islet xenotransplantation can restore the sensitivity of primate glucagon to insulin but cannot protect the diabetic recipient against hypoglycemia.
Collapse
Affiliation(s)
- Denis Dufrane
- Faculté de Médecine, Laboratory of Experimental Surgery, Université catholique de Louvain, CHEX 5570, Avenue Hippocrate 55, Brussels, Belgium.
| | | | | | | | | | | |
Collapse
|
143
|
Abstract
Glucokinase, a unique isoform of the hexokinase enzymes, which are known to phosphorylate D-glucose and other hexoses, was identified during the past three to four decades as a new, promising drug target for type 2 diabetes. Glucokinase serves as a glucose sensor of the insulin-producing pancreatic islet beta-cells, controls the conversion of glucose to glycogen in the liver and regulates hepatic glucose production. Guided by this fundamental knowledge, several glucokinase activators are now being developed, and have so far been shown to lower blood glucose in several animal models of type 2 diabetes and in initial trials in humans with the disease. Here, the scientific basis and current status of this new approach to diabetes therapy are discussed.
Collapse
|
144
|
Insulin signaling in alpha cells modulates glucagon secretion in vivo. Cell Metab 2009; 9:350-61. [PMID: 19356716 PMCID: PMC2694613 DOI: 10.1016/j.cmet.2009.02.007] [Citation(s) in RCA: 241] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2008] [Revised: 01/29/2009] [Accepted: 02/12/2009] [Indexed: 12/21/2022]
Abstract
Glucagon plays an important role in glucose homeostasis by regulating hepatic glucose output in both normo- and hypoglycemic conditions. In this study, we created and characterized alpha cell-specific insulin receptor knockout (alphaIRKO) mice to directly explore the role of insulin signaling in the regulation of glucagon secretion in vivo. Adult male alphaIRKO mice exhibited mild glucose intolerance, hyperglycemia, and hyperglucagonemia in the fed state and enhanced glucagon secretion in response to L-arginine stimulation. Hyperinsulinemic-hypoglycemic clamp studies revealed an enhanced glucagon secretory response and an abnormal norepinephrine response to hypoglycemia in alphaIRKO mice. The mutants also exhibited an age-dependent increase in beta cell mass. Furthermore, siRNA-mediated knockdown of insulin receptor in glucagon-secreting InR1G cells promoted enhanced glucagon secretion and complemented our in vivo findings. Together, these data indicate a significant role for intraislet insulin signaling in the regulation of alpha cell function in both normo- and hypoglycemic conditions.
Collapse
|
145
|
Abstract
Type 2 diabetes (T2DM) is not only a disorder of impaired insulin secretion but also glucagon oversecretion. However, the link between the two remains unclear. Is it possible that the latter is a consequence of the former? In this issue, Kawamori et al. (2009) have addressed this question by generating alpha cell-specific insulin receptor knockout mice.
Collapse
Affiliation(s)
- Jesper Gromada
- Novartis Institutes for BioMedical Research, 100 Technology Square, Cambridge, MA 02139, USA.
| | | | | |
Collapse
|
146
|
Hauge-Evans AC, King AJ, Carmignac D, Richardson CC, Robinson ICAF, Low MJ, Christie MR, Persaud SJ, Jones PM. Somatostatin secreted by islet delta-cells fulfills multiple roles as a paracrine regulator of islet function. Diabetes 2009; 58:403-11. [PMID: 18984743 PMCID: PMC2628614 DOI: 10.2337/db08-0792] [Citation(s) in RCA: 219] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
OBJECTIVE Somatostatin (SST) is secreted by islet delta-cells and by extraislet neuroendocrine cells. SST receptors have been identified on alpha- and beta-cells, and exogenous SST inhibits insulin and glucagon secretion, consistent with a role for SST in regulating alpha- and beta-cell function. However, the specific intraislet function of delta-cell SST remains uncertain. We have used Sst(-/-) mice to investigate the role of delta-cell SST in the regulation of insulin and glucagon secretion in vitro and in vivo. RESEARCH DESIGN AND METHODS Islet morphology was assessed by histological analysis. Hormone levels were measured by radioimmunoassay in control and Sst(-/-) mice in vivo and from isolated islets in vitro. RESULTS Islet size and organization did not differ between Sst(-/-) and control islets, nor did islet glucagon or insulin content. Sst(-/-) mice showed enhanced insulin and glucagon secretory responses in vivo. In vitro stimulus-induced insulin and glucagon secretion was enhanced from perifused Sst(-/-) islets compared with control islets and was inhibited by exogenous SST in Sst(-/-) but not control islets. No difference in the switch-off rate of glucose-stimulated insulin secretion was observed between genotypes, but the cholinergic agonist carbamylcholine enhanced glucose-induced insulin secretion to a lesser extent in Sst(-/-) islets compared with controls. Glucose suppressed glucagon secretion from control but not Sst(-/-) islets. CONCLUSIONS We suggest that delta-cell SST exerts a tonic inhibitory influence on insulin and glucagon secretion, which may facilitate the islet response to cholinergic activation. In addition, delta-cell SST is implicated in the nutrient-induced suppression of glucagon secretion.
Collapse
Affiliation(s)
- Astrid C Hauge-Evans
- Beta Cell Development and Function Group, Division of Reproduction and Endocrinology, King's College London, London, UK.
| | | | | | | | | | | | | | | | | |
Collapse
|
147
|
Quoix N, Cheng-Xue R, Mattart L, Zeinoun Z, Guiot Y, Beauvois MC, Henquin JC, Gilon P. Glucose and pharmacological modulators of ATP-sensitive K+ channels control [Ca2+]c by different mechanisms in isolated mouse alpha-cells. Diabetes 2009; 58:412-21. [PMID: 19008345 PMCID: PMC2628615 DOI: 10.2337/db07-1298] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [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 We studied how glucose and ATP-sensitive K(+) (K(ATP)) channel modulators affect alpha-cell [Ca(2+)](c). RESEARCH DESIGN AND METHODS GYY mice (expressing enhanced yellow fluorescent protein in alpha-cells) and NMRI mice were used. [Ca(2+)](c), the K(ATP) current (I(KATP), perforated mode) and cell metabolism [NAD(P)H fluorescence] were monitored in single alpha-cells and, for comparison, in single beta-cells. RESULTS In 0.5 mmol/l glucose, [Ca(2+)](c) oscillated in some alpha-cells and was basal in the others. Increasing glucose to 15 mmol/l decreased [Ca(2+)](c) by approximately 30% in oscillating cells and was ineffective in the others. alpha-Cell I(KATP) was inhibited by tolbutamide and activated by diazoxide or the mitochondrial poison azide, as in beta-cells. Tolbutamide increased alpha-cell [Ca(2+)](c), whereas diazoxide and azide abolished [Ca(2+)](c) oscillations. Increasing glucose from 0.5 to 15 mmol/l did not change I(KATP) and NAD(P)H fluorescence in alpha-cells in contrast to beta-cells. The use of nimodipine showed that L-type Ca(2+) channels are the main conduits for Ca(2+) influx in alpha-cells. gamma-Aminobutyric acid and zinc did not decrease alpha-cell [Ca(2+)](c), and insulin, although lowering [Ca(2+)](c) very modestly, did not affect glucagon secretion. CONCLUSIONS alpha-Cells display similarities with beta-cells: K(ATP) channels control Ca(2+) influx mainly through L-type Ca(2+) channels. However, alpha-cells have distinct features from beta-cells: Most K(ATP) channels are already closed at low glucose, glucose does not affect cell metabolism and I(KATP), and it slightly decreases [Ca(2+)](c). Hence, glucose and K(ATP) channel modulators exert distinct effects on alpha-cell [Ca(2+)](c). The direct small glucose-induced drop in alpha-cell [Ca(2+)](c) contributes likely only partly to the strong glucose-induced inhibition of glucagon secretion in islets.
Collapse
Affiliation(s)
- Nicolas Quoix
- Unit of Endocrinology and Metabolism, University of Louvain Faculty of Medicine, Brussels, Belgium
| | | | | | | | | | | | | | | |
Collapse
|
148
|
Affiliation(s)
- David A Jacobson
- Department of Medicine, Section of Endocrinology, Diabetes and Metabolism, The University of Chicago, Chicago, Illinois, USA
| | | | | |
Collapse
|
149
|
Affiliation(s)
- Guy A Rutter
- Department of Cell Biology, Division of Medicine, Faculty of Medicine, Imperial College London, London, UK.
| |
Collapse
|
150
|
Beneficial effects of intercellular interactions between pancreatic islet cells in blood glucose regulation. J Theor Biol 2008; 257:312-9. [PMID: 19135066 DOI: 10.1016/j.jtbi.2008.12.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2008] [Revised: 11/26/2008] [Accepted: 12/03/2008] [Indexed: 11/20/2022]
Abstract
Glucose homeostasis is controlled by the islets of Langerhans which are equipped with alpha-cells increasing the blood glucose level, beta-cells decreasing it, and delta-cells the precise role of which still needs identifying. Although intercellular communications between these endocrine cells have recently been observed, their roles in glucose homeostasis have not been clearly understood. In this study, we construct a mathematical model for an islet consisting of two-state alpha-, beta-, and delta-cells, and analyze effects of known chemical interactions between them with emphasis on the combined effects of those interactions. In particular, such features as paracrine signals of neighboring cells and cell-to-cell variations in response to external glucose concentrations as well as glucose dynamics, depending on insulin and glucagon hormone, are considered explicitly. Our model predicts three possible benefits of the cell-to-cell interactions: First, the asymmetric interaction between alpha- and beta-cells contributes to the dynamic stability while the perturbed glucose level recovers to the normal level. Second, the inhibitory interactions of delta-cells for glucagon and insulin secretion prevent the wasteful co-secretion of them at the normal glucose level. Finally, the glucose dose-responses of insulin secretion is modified to become more pronounced at high glucose levels due to the inhibition by delta-cells. It is thus concluded that the intercellular communications in islets of Langerhans should contribute to the effective control of glucose homeostasis.
Collapse
|