Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 229
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Am J Transplant ; 13(6): 1461-73, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23679575

ABSTRACT

Keratoplasty is the primary treatment to cure blindness due to corneal opacification. However, immune-mediated rejection remains the leading cause of keratoplasty failure. Here, we utilize an in vivo imaging approach to monitor, track, and characterize in real-time the recruitment of GFP-labeled allo-specific activated (Bonzo) T cells during corneal allograft rejection. We show that the recruitment of effector T cells to the site of transplantation determined the fate of corneal allografts, and that local intra-graft production of CCL5 and CXCL9/10 regulated motility patterns of effector T cells in situ, and correlated with allograft rejection. We also show that different motility patterns associate with distinct in vivo phenotypes (round, elongated, and ruffled) of graft-infiltrating effector T cells with varying proportions during progression of rejection. The ruffled phenotype was characteristic of activated effectors T cells and predominated during ongoing rejection, which associated with significantly increased T cell dynamics within the allografts. Importantly, CCR5/CXCR3 blockade decreased the motility, size, and number of infiltrating T cells and significantly prolonged allograft survival. Our findings indicate that chemokines produced locally within corneal allografts play an important role in the in situ activation and dynamic behavior of infiltrating effector T cells, and may guide targeted interventions to promote graft survival.


Subject(s)
Cell Movement/immunology , Chemokines/immunology , Cornea/immunology , Corneal Transplantation , Graft Rejection/immunology , Immunity, Cellular , T-Lymphocytes/pathology , Animals , Chemokines/metabolism , Cornea/pathology , Corneal Diseases/surgery , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Graft Rejection/pathology , Graft Survival , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , T-Lymphocytes/immunology , Transplantation, Homologous
2.
Diabetes Obes Metab ; 15 Suppl 3: 105-16, 2013 Sep.
Article in English | MEDLINE | ID: mdl-24003927

ABSTRACT

Although, diabetes is reaching pandemic proportions, the exact aetiology of either type 1 (T1D) or type 2 diabetes (T2D) remains to be determined. Mounting evidence, however, suggests that islet inflammation is a likely common denominator during early development of either type of the disease. In this review, we highlight some of the inflammatory mechanisms that appear to be shared between T1D and T2D, and we explore the utility of intravital imaging in the study of islet inflammation. Intravital imaging has emerged as an indispensable tool in biomedical research and a variety of in vivo imaging approaches have been developed to study pancreatic islet physiology and pathophysiology in the native environment in health and disease. However, given the scattered distribution of the islets of Langerhans within the 'sea' of the exocrine pancreas located deep within the body and the fact that the islets only constitute 1-2% of the total volume of pancreatic tissue, studying the pancreatic islet in situ has been challenging. Here, we focus on a new experimental approach that enables studying local islet inflammation with single-cell resolution in the relevant context of the in vivo environment non-invasively and longitudinally and, thereby improving our understanding of diabetes pathogenesis.


Subject(s)
Cell Tracking/methods , Diagnostic Imaging/methods , Inflammation/diagnosis , Islets of Langerhans/pathology , Animals , Diabetes Mellitus/diagnosis , Diabetes Mellitus/etiology , Diabetes Mellitus/pathology , Humans , Injections, Intraocular , Islets of Langerhans Transplantation , Microscopy, Confocal , Pancreatitis/diagnosis , Pancreatitis/pathology
3.
Nat Genet ; 26(3): 336-40, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11062475

ABSTRACT

Mitochondrial dysfunction is an important contributor to human pathology and it is estimated that mutations of mitochondrial DNA (mtDNA) cause approximately 0.5-1% of all types of diabetes mellitus. We have generated a mouse model for mitochondrial diabetes by tissue-specific disruption of the nuclear gene encoding mitochondrial transcription factor A (Tfam, previously mtTFA; ref. 7) in pancreatic beta-cells. This transcriptional activator is imported to mitochondria, where it is essential for mtDNA expression and maintenance. The Tfam-mutant mice developed diabetes from the age of approximately 5 weeks and displayed severe mtDNA depletion, deficient oxidative phosphorylation and abnormal appearing mitochondria in islets at the ages of 7-9 weeks. We performed physiological studies of beta-cell stimulus-secretion coupling in islets isolated from 7-9-week-old mutant mice and found reduced hyperpolarization of the mitochondrial membrane potential, impaired Ca(2+)-signalling and lowered insulin release in response to glucose stimulation. We observed reduced beta-cell mass in older mutants. Our findings identify two phases in the pathogenesis of mitochondrial diabetes; mutant beta-cells initially display reduced stimulus-secretion coupling, later followed by beta-cell loss. This animal model reproduces the beta-cell pathology of human mitochondrial diabetes and provides genetic evidence for a critical role of the respiratory chain in insulin secretion.


Subject(s)
DNA, Mitochondrial/analysis , DNA-Binding Proteins , Diabetes Mellitus/genetics , Disease Models, Animal , High Mobility Group Proteins , Insulin/metabolism , Islets of Langerhans/pathology , Mitochondrial Proteins , Nuclear Proteins , Trans-Activators , Transcription Factors/deficiency , Viral Proteins , Xenopus Proteins , Adenosine Triphosphate/metabolism , Age Factors , Animals , Calcium/pharmacology , Calcium Channels, L-Type/metabolism , Diabetes Mellitus/pathology , Disease Progression , Electron Transport Complex IV/analysis , Exocytosis , Gene Targeting , Glucose/pharmacology , Humans , Insulin Secretion , Integrases/metabolism , Ion Transport , Islets of Langerhans/metabolism , Mice , Mice, Transgenic , Organ Specificity , Oxidative Phosphorylation , Potassium Channels/metabolism , Recombinant Fusion Proteins/metabolism , Secretory Rate , Succinate Dehydrogenase/analysis , Transcription Factors/genetics , Transcription Factors/physiology , Transcription, Genetic , Transgenes
4.
Diabetologia ; 54(5): 1121-6, 2011 May.
Article in English | MEDLINE | ID: mdl-21360190

ABSTRACT

AIMS/HYPOTHESIS: The aim of this study was to provide evidence that the anterior chamber of the eye serves as a novel clinical islet implantation site. METHODS: In a preclinical model, allogeneic pancreatic islets were transplanted into the anterior chamber of the eye of a baboon model for diabetes, and metabolic and ophthalmological outcomes were assessed. RESULTS: Islets readily engrafted on the iris and there was a decrease in exogenous insulin requirements due to insulin secretion from the intraocular grafts. No major adverse effects on eye structure and function could be observed during the transplantation period. CONCLUSIONS/INTERPRETATION: Our study demonstrates the long-term survival and function of allogeneic islets after transplantation into the anterior chamber of the eye. The safety and simplicity of this procedure provides support for further studies aimed at translating this technology into the clinic.


Subject(s)
Anterior Chamber/surgery , Diabetes Mellitus, Experimental/therapy , Islets of Langerhans Transplantation/methods , Animals , Papio
5.
Article in English | MEDLINE | ID: mdl-33816651

ABSTRACT

The islets of Langerhans constitute the endocrine pancreas which regulates blood glucose homeostasis and their dysfunction results in diabetes. Each of the pancreatic islets constitutes an entire micro-organ with intricate cell to cell interactions and that is well vascularized and innervated. An important therapeutic advantage in islet transplant is that pancreatic islets maintain their organ integrity when isolated and transplanted to patients with severe diabetes. Once transplanted, the islet micro-organs actively contribute to their own vascularization and start to function immediately. Hence, in terms of organ transplantation, the application of pancreatic islets will be a decisive clinical tool for future diabetes care (credit: Tilo Moede).

6.
Science ; 278(5337): 471-4, 1997 Oct 17.
Article in English | MEDLINE | ID: mdl-9334307

ABSTRACT

Inositol hexakisphosphate (InsP6), the dominant inositol phosphate in insulin-secreting pancreatic beta cells, inhibited the serine-threonine protein phosphatases type 1, type 2A, and type 3 in a concentration-dependent manner. The activity of voltage-gated L-type calcium channels is increased in cells treated with inhibitors of serine-threonine protein phosphatases. Thus, the increased calcium channel activity obtained in the presence of InsP6 might result from the inhibition of phosphatase activity. Glucose elicited a transient increase in InsP6 concentration, which indicates that this inositol polyphosphate may modulate calcium influx over the plasma membrane and serve as a signal in the pancreatic beta cell stimulus-secretion coupling.


Subject(s)
Calcium Channels/metabolism , Islets of Langerhans/metabolism , Phosphoprotein Phosphatases/antagonists & inhibitors , Phytic Acid/pharmacology , Animals , Calcium/metabolism , Calcium Channels/drug effects , Cell Membrane/metabolism , Cricetinae , Dose-Response Relationship, Drug , Glucose/pharmacology , Inositol/pharmacology , Inositol Phosphates/pharmacology , Ion Channel Gating , Islets of Langerhans/drug effects , Patch-Clamp Techniques , Tumor Cells, Cultured
7.
Science ; 271(5250): 813-5, 1996 Feb 09.
Article in English | MEDLINE | ID: mdl-8628999

ABSTRACT

Hypoglycemic sulfonylureas represent a group of clinically useful antidiabetic compounds that stimulate insulin secretion from pancreatic beta cells. The molecular mechanisms involved are not fully understood but are believed to involve inhibition of potassium channels sensitive to adenosine triphosphate (KATP channels) in the beta cell membrane, causing membrane depolarization, calcium influx, and activation of the secretory machinery. In addition to these effects, sulfonylureas also promoted exocytosis by direct interaction with the secretory machinery not involving closure of the plasma membrane KATP channels. This effect was dependent on protein kinase C (PKC) and was observed at therapeutic concentrations of sulfonylureas, which suggests that it contributes to their hypoglycemic action in diabetics.


Subject(s)
Exocytosis/drug effects , Hypoglycemic Agents/pharmacology , Islets of Langerhans/physiology , Protein Kinase C/metabolism , Sulfonylurea Compounds/pharmacology , Tolbutamide/pharmacology , Animals , Calcium/metabolism , Cells, Cultured , Cytoplasmic Granules/metabolism , Electric Conductivity , Glipizide/pharmacology , Glyburide/pharmacology , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/drug effects , Membrane Potentials/drug effects , Mice , Patch-Clamp Techniques
8.
J Clin Invest ; 92(4): 1910-7, 1993 Oct.
Article in English | MEDLINE | ID: mdl-8408643

ABSTRACT

To extend previous observations on the role of polyamines in insulin production, metabolism, and replication of insulin-secreting pancreatic beta cells, we have studied the role of polyamines in the regulation of the stimulus-secretion coupling of clonal rat insulinoma cells (RINm5F). For this purpose, RINm5F cells were partially depleted in their polyamine contents by use of the specific ornithine decarboxylase inhibitor difluoromethylornithine (DFMO), which led to an increase in cellular insulin and ATP contents. Analysis of different parts of the signal transduction pathway revealed that insulin secretion and the increase in cytoplasmic free Ca2+ concentration ([Ca2+]i) after K(+)-induced depolarization were markedly enhanced in DFMO-treated cells. These effects were paralleled by increased voltage-activated Ca2+ currents, as judged by whole-cell patch-clamp analysis, probably reflecting increased channel activity rather than elevated number of channels per cell. DFMO treatment also rendered phospholipase C in these cells more sensitive to the muscarinic receptor agonist carbamylcholine, as evidenced by enhanced generation of inositol phosphates, increase in [Ca2+]i and insulin secretion, despite an unaltered ligand binding to muscarinic receptors and lack of effect on protein kinase C activity. In addition, the tumor promoter 12-O-tetradecanoylphorbol 13-acetate, at concentrations suggested to be specific for protein kinase C activation, evoked an increased insulin output in polyamine-deprived cells compared to control cells. The stimulatory effects of glucose or the cyclic AMP raising agent theophylline on insulin release were not increased by DFMO treatment. In spite of increased binding of sulfonylurea in DFMO-treated cells, there was no secretory response or altered increase in [Ca2+]i in response to the drug in these cells. It is concluded that partial polyamine depletion sensitizes the stimulus-secretion coupling at multiple levels in the insulinoma cells, including increased voltage-dependent Ca2+ influx and enhanced responsiveness to activators of phospholipase C and protein kinase C. In their entirety, our present results indicate that the behavior of the stimulus-secretion coupling of polyamine-depleted RINm5F insulinoma cells changes towards that of native beta cells, thus improving the usefulness of this cell line for studies of beta cell insulin secretion.


Subject(s)
Calcium/metabolism , Eflornithine/pharmacology , Inositol Phosphates/metabolism , Insulin/metabolism , Insulinoma/metabolism , Pancreatic Neoplasms/metabolism , Polyamines/metabolism , Adenosine Triphosphate/metabolism , Animals , Calcium Channels/biosynthesis , Calcium Channels/drug effects , Calcium Channels/physiology , Carbachol/pharmacology , Cytoplasm/drug effects , Cytoplasm/metabolism , DNA, Neoplasm/metabolism , Diazoxide/pharmacology , Glucose/pharmacology , Glyburide/pharmacology , Insulin Secretion , Kinetics , Potassium/pharmacology , Putrescine/metabolism , RNA, Messenger/metabolism , Rats , Spermidine/metabolism , Spermine/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Theophylline/pharmacology , Tolbutamide/pharmacology , Tumor Cells, Cultured
9.
Biosci Rep ; 27(6): 321-6, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17597394

ABSTRACT

The aim of this study was to clarify the frequency of patients with type 1 diabetes that have serum that increases pancreatic beta-cell cytoplasmic free Ca(2+) concentration, [Ca(2+)](i), and if such an effect is also present in serum from first-degree relatives. We also studied a possible link between the serum effect and ethnic background as well as presence of autoantibodies. Sera obtained from three different countries were investigated as follows: 82 Swedish Caucasians with newly diagnosed type 1 diabetes, 56 Americans with different duration of type 1 diabetes, 117 American first-degree relatives of type 1 diabetic patients with a mixed ethnic background and 31 Caucasian Finnish children with newly diagnosed type 1 diabetes. Changes in [Ca(2+)](i) , upon depolarization, were measured in beta-cells incubated overnight with sera from type 1 diabetic patients, first-degree relatives or healthy controls. Our data show that there is a group constituting approximately 30% of type 1 diabetic patients of different gender, age, ethnic background and duration of the disease, as well as first-degree relatives of type 1 diabetic patients, that have sera that interfere with pancreatic beta-cell Ca(2+)-handling. This effect on beta-cell [Ca(2+)](i) could not be correlated to the presence of autoantibodies. In a defined subgroup of patients with type 1 diabetes and first-degree relatives a defect Ca(2+)-handling may aggravate development of beta-cell destruction.


Subject(s)
Calcium/metabolism , Diabetes Mellitus, Type 1/blood , Insulin-Secreting Cells/metabolism , Serum/metabolism , Adolescent , Adult , Age Factors , Animals , Autoantibodies/blood , Autoantibodies/immunology , Cells, Cultured , Child , Child, Preschool , Diabetes Mellitus, Type 1/ethnology , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Family , Female , Finland , Humans , Infant , Insulin-Secreting Cells/pathology , Male , Mice , Serum/immunology , Sex Factors , Sweden , Time Factors , United States
10.
Mol Biol Cell ; 9(5): 1209-20, 1998 May.
Article in English | MEDLINE | ID: mdl-9571250

ABSTRACT

Dopamine (DA) inhibition of Na+,K+-ATPase in proximal tubule cells is associated with increased endocytosis of its alpha and beta subunits into early and late endosomes via a clathrin vesicle-dependent pathway. In this report we evaluated intracellular signals that could trigger this mechanism, specifically the role of phosphatidylinositol 3-kinase (PI 3-K), the activation of which initiates vesicular trafficking and targeting of proteins to specific cell compartments. DA stimulated PI 3-K activity in a time- and dose-dependent manner, and this effect was markedly blunted by wortmannin and LY 294002. Endocytosis of the Na+,K+-ATPase alpha subunit in response to DA was also inhibited in dose-dependent manner by wortmannin and LY 294002. Activation of PI 3-K generally occurs by association with tyrosine kinase receptors. However, in this study immunoprecipitation with a phosphotyrosine antibody did not reveal PI 3-K activity. DA-stimulated endocytosis of Na+, K+-ATPase alpha subunits required protein kinase C, and the ability of DA to stimulate PI 3-K was blocked by specific protein kinase C inhibitors. Activation of PI 3-K is mediated via the D1 receptor subtype and the sequential activation of phospholipase A2, arachidonic acid, and protein kinase C. The results indicate a key role for activation of PI 3-K in the endocytic sequence that leads to internalization of Na+,K+-ATPase alpha subunits in response to DA, and suggest a mechanism for the participation of protein kinase C in this process.


Subject(s)
Dopamine/pharmacology , Endocytosis/physiology , Kidney Cortex/enzymology , Phosphatidylinositol 3-Kinases/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Cells, Cultured , Enzyme Activation , Male , Rats , Rats, Sprague-Dawley
11.
Article in English | MEDLINE | ID: mdl-29497631

ABSTRACT

It is now well established that beta cell replacement through pancreatic islet transplantation results in significant improvement in the quality-of-life of type 1 diabetes (T1D) patients. This is achieved through improved control and prevention of severe drops in blood sugar levels. Islet transplant therapy is on the verge of becoming standard-of-care in the USA. Yet, as with other established transplantation therapies, there remain hurdles to overcome to bring islet transplantation to full fruition as a long-lasting therapy of T1D. One of these hurdles is establishing reliable new sites, other than the liver, where durable efficacy and survival of transplanted islets can be achieved. In this article, we discuss the anterior chamber of the eye as a new site for clinical islet transplantation in the treatment of T1D. We specifically focus on the common conceptions, and preconceptions, on the requirements of islet mass, and whether or not the anterior chamber can accommodate sufficient islets to achieve meaningful efficacy and significant impact on hyperglycemia in clinical application.

12.
Biochim Biophys Acta ; 887(2): 236-41, 1986 Jul 11.
Article in English | MEDLINE | ID: mdl-3521749

ABSTRACT

The effects of 12-O-tetradecanoylphorbol 13-acetate (TPA) on the handling of Ca2+ and insulin release were investigated in the clonal insulin-producing cell line RINm5F. The presence of the phorbol ester lowered the free cytoplasmic Ca2+ and suppressed the increase obtained by depolarization with high concentrations of K+. Despite the lowering in cytoplasmic Ca2+ by TPA, there was a concomitant stimulation of insulin release indicating that one feature of protein kinase C activation is to make the secretory system more sensitive to Ca2+. Furthermore, there was no interaction of TPA with the mechanisms responsible for inositol 1,4,5-tris(phosphate) induced Ca2+ release or Ca2+ uptake in permeabilized cells. Although TPA slightly depolarized the RINm5F cells there was no interference with K+-induced depolarization. It is suggested that an additional effect of protein kinase C activation in these cells, is to stimulate the extrusion of Ca2+ over the plasma membrane.


Subject(s)
Calcium/physiology , Insulin/metabolism , Phorbols/pharmacology , Tetradecanoylphorbol Acetate/pharmacology , Animals , Cell Line , Cytoplasm/metabolism , Inositol Phosphates/metabolism , Insulin Secretion , Membrane Potentials/drug effects , Protein Kinase C/metabolism , Rats , Secretory Rate/drug effects
13.
Biochim Biophys Acta ; 720(3): 320-8, 1982 Jun 08.
Article in English | MEDLINE | ID: mdl-6285995

ABSTRACT

Electrothermal atomic absorption spectroscopy was employed for measuring barium in beta-cell-rich pancreatic islets microdissected from ob/ob-mice. Both the uptake and efflux of barium displayed two distinct phases. There was a 4-fold accumulation of barium into intracellular stores when its extracellular concentration was 0.26 mM. Unlike divalent cations with more extensive intracellular accumulation, the washout of Ba2+ was not inhibited by D-glucose. Ba2+ served as a substitute for Ca2+ both in maintaining the glucose metabolism after removal of extracellular Ca2+ and making it possible for glucose to stimulate insulin release. Furthermore, Ba2+ elicited insulin release in the absence of glucose and other secretagogues. The latter effect was reversible and was markedly potentiated under conditions known to increase the beta-cell content of cyclic AMP. It is likely that the observed actions of Ba2+ are mediated by Ca2+, since Ca2+ -dependent regulatory proteins, such as calmodulin, apparently cannot bind Ba2+ specifically.


Subject(s)
Barium/metabolism , Insulin/metabolism , Islets of Langerhans/metabolism , Animals , Biological Transport , Calcium/physiology , Cyclic AMP/metabolism , Glucose/metabolism , Insulin Secretion , Mice , Mice, Obese , Oxidation-Reduction
14.
Biochim Biophys Acta ; 929(1): 81-7, 1987 Jun 15.
Article in English | MEDLINE | ID: mdl-3297172

ABSTRACT

A possible interaction between Cd2+ and Ca2+ as a component in Cd2+-induced insulin release was investigated in beta cells isolated from obese hyperglycemic mice. The glucose stimulated Cd2+ uptake was dependent on the concentration of sugar. This uptake was sigmoidal with a Km for glucose of about 5 mM and was suppressed by both 50 microM of the voltage-activated Ca2+ channel blocker D-600 and 12 mM Mg2+. In the presence of 8 mM glucose 5 microM Cd2+ evoked a prompt and sustained stimulatory response, corresponding to about 3-fold of the insulin release obtained in the absence of the ion. Whereas 5 microM Cd2+ was without effect on the glucose-stimulated 45Ca efflux in the presence of extracellular Ca2+, 40 microM inhibited it. At a concentration of 5 microM, Cd2+ had no effect on the resting membrane potential or the depolarization evoked by either glucose or K+. In the absence of extracellular Ca2+ there was only a modest stimulation of 45Ca efflux by 5 microM Cd2+. Studies of the ambient free Ca2+ concentration maintained by permeabilized cells also indicate that 5 microM Cd2+ do not mobilize intracellularly bound Ca2+ to any great extent. On the contrary, at this concentration, Cd2+ even suppressed inositol 1,4,5-trisphosphate (IP3)-induced Ca2+ release. The present study suggests that Cd2+ stimulates insulin release by a direct mechanism which does not involve an increase in cytoplasmic free Ca2+ concentration.


Subject(s)
Cadmium/pharmacology , Calcium/metabolism , Insulin/metabolism , Islets of Langerhans/drug effects , Animals , Calcium/pharmacology , Drug Interactions , Female , Glucose/pharmacology , Hyperglycemia/metabolism , Inositol Phosphates/metabolism , Insulin Secretion , Intracellular Fluid/metabolism , Islets of Langerhans/metabolism , Male , Mice , Mice, Obese/metabolism
15.
Biochim Biophys Acta ; 888(3): 270-7, 1986 Oct 10.
Article in English | MEDLINE | ID: mdl-3530337

ABSTRACT

The transport of Cd2+ and the effects of this ion on secretory activity and metabolism were investigated in beta cell-rich pancreatic islets isolated from obese-hyperglycemic mice. The endogenous cadmium content was 2.5 mumol/kg dry wt. After 60 min of incubation in a Ca2+-deficient medium containing 2.5 microM Cd2+ the islet cadmium content increased to 0.18 mmol/kg dry wt. This uptake was reduced by approx. 50% in the presence of 1.28 mM Ca2+. The incorporation of Cd2+ was stimulated either by raising the concentration of glucose to 20 mM or K+ to 30.9 mM. Whereas D-600 suppressed the stimulatory effect of glucose by 75%, it completely abolished that obtained with high K+. Only about 40% of the incorporated cadmium was mobilized during 60 min of incubation in a Cd2+-free medium containing 0.5 mM EGTA. It was possible to demonstrate a glucose-induced suppression of Cd2+ efflux into a Ca2+-deficient medium. Concentrations of Cd2+ up to 2.5 microM did not affect glucose oxidation, whereas, there was a progressive inhibition when the Cd2+ concentration was above 10 microM. Basal insulin release was stimulated by 5 microM Cd2+. At a concentration of 160 microM, Cd2+ did not affect basal insulin release but significantly inhibited the secretory response to glucose. It is concluded that the beta cell uptake of Cd2+ is facilitated by the activation of voltage-dependent Ca2+ channels. Apparently, the accumulation of Cd2+ mimics that of Ca2+ also involving a component of intracellular sequestration promoted by glucose.


Subject(s)
Cadmium/metabolism , Calcium/metabolism , Glucose/pharmacology , Islets of Langerhans/metabolism , Potassium/pharmacology , Animals , Cadmium/analysis , Glucose/metabolism , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/analysis , Islets of Langerhans/drug effects , Mice , Mice, Obese , Pancreas/analysis
16.
Biochim Biophys Acta ; 1223(2): 226-33, 1994 Sep 08.
Article in English | MEDLINE | ID: mdl-8086492

ABSTRACT

The plasma membrane Ca2+ carrier system involved in receptor-mediated Ca2+ entry was studied. Using the Ca2+ readdition protocol, the rate of cytosolic free Ca2+ concentration ([Ca2+]i) increase in vasopressin-pretreated hepatocytes was significantly higher than in thapsigargin- or 2,5-di(tert-butyl)hydroquinone-pretreated cells. The addition of Mn2+ to unstimulated hepatocytes resulted in a biphasic quench of fura-2 fluorescence. After an initial phase that was fast in rate but of short duration, the rate of fura-2 quench by Mn2+ became much slower and lasted until all the cellular fura-2 was quenched. Pretreatment of the cells with vasopressin only accelerated the rate of the latter phase but not of the initial one. In agonist-stimulated cells, acidification of the extracellular medium or the presence of ruthenium red, econazole or SK&F 96365 decreased the rates of both [Ca2+]i increase and Mn2+ entry upon addition of the respective cation. By contrast, neomycin and N-tosyl-L-phenylalanine chloromethyl ketone markedly decreased the rate of [Ca2+]i increase upon Ca2+ readdition but had no effect on vasopressin-stimulated Mn2+ entry. None of the treatments affected the ability of vasopressin and thapsigargin to mobilize the internal Ca2+ store. It is concluded that in hepatocytes the two pathways of receptor-mediated Ca2+ entry control two distinct yet pharmacologically related cation carriers.


Subject(s)
Calcium-Binding Proteins/metabolism , Calcium/metabolism , Receptors, Cell Surface/metabolism , Animals , Calcium/antagonists & inhibitors , Calcium Channels/metabolism , Calcium-Binding Proteins/antagonists & inhibitors , Cell Membrane/metabolism , Culture Media , Imidazoles/pharmacology , Liver/metabolism , Male , Rats , Rats, Wistar , Terpenes/pharmacology , Thapsigargin , Type C Phospholipases/metabolism , Vasopressins/pharmacology
17.
Diabetes ; 45(5): 580-6, 1996 May.
Article in English | MEDLINE | ID: mdl-8621007

ABSTRACT

We studied the effects of fatty acid oxidation on insulin secretion of db/db mice and underlying molecular mechanisms of these effects. At 2-3 months of age, db/db mice were markedly obese, hyperglycemic, and hyperinsulinemic. Serum free fatty acid (FFA) levels were increased in 2-month-old (1.5 +/- 0.1 vs. 1.1 +/- 0.1 mmol/l, P < 0.05) and 3-month-old (1.9 +/- 0.1 vs. 1.2 +/- 0.1 mmol/l, P < 0.01) mice compared with the age and sex-matched db/+ mice serving as controls. Glucose-induced insulin release from db/db islets was markedly decreased compared with that from db/+ islets and was specifically ameliorated (by 54% in 2-month-old and 38% in 3-month-old mice) by exposure to a carnitine palmitoyltransferase I inhibitor, etomoxir (1 micromol/l). Etomoxir failed to affect the insulin response to alpha-ketoisocaproate. The effect of etomoxir on glucose-induced insulin release was lost after culturing db/db islets in RPMI medium containing 22 mmol/l glucose but no fatty acid. Culture of db/+ islets with 0.125 mmol/l palmitate led to a decrease in glucose-induced insulin secretion, which was partially reversible by etomoxir. Both islet glucose oxidation and the ratio of glucose oxidation to utilization were decreased in db/db islets. Etomoxir significantly enhanced glucose oxidation by 60% and also the ratio of oxidation to glucose utilization (from 27 +/- 2.5 to 37 +/-3.0%, P < 0.05). Pyruvate dehydrogenase (PDH) activity was decreased in islets of db/db mice (75 +/-4.2 vs. 91 +/- 2.9 nU/ng DNA, P < 0.01), whereas PDH kinase activity was increased (rate of PDH inactivation -0.25 +/- 0.02 vs. - 0.11 +/- 0.02/min, P < 0.0 1). These abnormalities were partly but not wholly reversed by a 2-h preexposure to etomoxir. In conclusion, elevated FFA levels in the db/db mouse diminish glucose-induced insulin secretion by a glucose-fatty acid cycle in which fatty acid oxidation inhibits glucose oxidation by decreasing PDH activity and increasing PDH kinase activities.


Subject(s)
Diabetes Mellitus/physiopathology , Enzyme Inhibitors/pharmacology , Epoxy Compounds/pharmacology , Fatty Acids, Nonesterified/blood , Hyperglycemia/physiopathology , Insulin/metabolism , Islets of Langerhans/physiopathology , Mitochondria/enzymology , Obesity/physiopathology , Pyruvate Dehydrogenase Complex/metabolism , Aging , Animals , Carnitine O-Palmitoyltransferase/antagonists & inhibitors , Cells, Cultured , Crosses, Genetic , DNA/analysis , Diabetes Mellitus/blood , Female , Glucose/pharmacology , Hyperglycemia/blood , Hyperinsulinism , Insulin/blood , Insulin Secretion , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Keto Acids/pharmacology , Kinetics , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/blood
18.
Diabetes ; 49(9): 1409-12, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10969822

ABSTRACT

Phosphatidylinositol 4,5-bisphosphate (PIP2) has been suggested to play an important role as an endogenous regulator of ATP-sensitive potassium (KATP) channels consisting of Kir6.2 as a pore-forming subunit. These studies show the ability of PIP2 to activate KATP channel activity and to counteract the inhibitory effect of ATP, implying that PIP2 could serve the function of modulating the sensitivity of KATP channels to the cytoplasmic free ATP concentration. Careful examination of the literature reveals that the definitive physiologically relevant experiments to establish efficacy of PIP2 on this channel may still have to be performed. Our reservations are based on the handling of PIP2 in cell-free experiments and in various strategies designed to modulate PIP2 concentrations in intact cells. Furthermore, a potent stimulatory effect of phosphatidylinositol 3,4,5trisphosphate, a downstream metabolite of PIP2, on KATP channel activity raises the possibility that the effects on the KATP channel may not be directly related to PIP2.


Subject(s)
Adenosine Triphosphate/physiology , Islets of Langerhans/physiology , Phosphatidylinositol 4,5-Diphosphate/physiology , Potassium Channels, Inwardly Rectifying , Potassium Channels/physiology , Adenosine Triphosphate/pharmacology , Animals , Cells, Cultured , Cytoplasm/physiology , Female , Islets of Langerhans/drug effects , Male , Mice , Mice, Obese , Patch-Clamp Techniques , Phosphatidylinositol 4,5-Diphosphate/pharmacology , Potassium Channels/drug effects
19.
Diabetes ; 50 Suppl 1: S15-9, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11272178

ABSTRACT

Fluorescent proteins have been extensively used as protein "tags" to study the subcellular localization of proteins and/or their translocation upon stimulation or as markers for transfection in transient and stable expression systems. However, they have not been frequently used as reporter genes to monitor stimulus-induced gene expression in mammalian cells. Here we demonstrate the use of fluorescent proteins to study stimulus-induced gene transcription. The general applicability of the approach is exemplified by doxycyclin-(Tet-On) and phorbol 12-myristate 13-acetate-induced (c-fos) promoter activation, with green fluorescent protein (GFP) and red fluorescent protein (DsRed) as semiquantitative and immediate reporters, of transcription activation. Under the control of beta-cell-specific promoters, such as the rat insulin 1 promoter or the rat upstream glucokinase promoter, this approach allowed us to monitor online glucose-induced gene transcription in primary beta-cells at the single-cell level as well as in the context of the islet of Langerhans. Applying discretely detectable fluorescent proteins, for example GFP and DsRed, enabled us to simultaneously monitor stimulus-induced transcription by two different promoters in the same cell.


Subject(s)
Glucose/pharmacology , Insulin/pharmacology , Islets of Langerhans/drug effects , Animals , Cell Line , Gene Expression Regulation/drug effects , Genes, Reporter/genetics , Green Fluorescent Proteins , Image Processing, Computer-Assisted , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Luminescent Proteins/drug effects , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Microscopy, Confocal , Microscopy, Fluorescence , Promoter Regions, Genetic/genetics , Recombinant Fusion Proteins/drug effects , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Time Factors
20.
Diabetes ; 50 Suppl 1: S44-7, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11272200

ABSTRACT

Apoptosis is a physiological form of cell death that occurs during normal development, and critical mediators of this process include caspases, reactive oxygen species, and Ca2+. Excessive apoptosis of the pancreatic beta-cell has been associated with diabetes. Consequently, apoptosis research has focused on how infiltrating macrophages or cytotoxic T-cells might kill pancreatic beta-cells using cytokines or death receptor triggering. Meanwhile, the intracellular events in the target beta-cell have been largely ignored. Elucidation of such targets might help develop improved treatment strategies for diabetes. This article will outline recent developments in apoptosis research, with emphasis on mechanisms that may be relevant to beta-cell death in type 1 and type 2 diabetes. Several of the models proposed in beta-cell killing converge on Ca2+ signaling, indicating that the pancreatic beta-cell may be an ideal system in which to carefully dissect the role of Ca2+ during apoptosis.


Subject(s)
Apoptosis/physiology , Cell Death/physiology , Diabetes Mellitus/physiopathology , Islets of Langerhans/cytology , Animals , Calcium/metabolism , Caspases/metabolism , Diabetes Mellitus/pathology , Humans , Islets of Langerhans/metabolism , Models, Biological , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL