Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Diabetes ; 70(1): 111-118, 2021 01.
Article in English | MEDLINE | ID: mdl-32826297

ABSTRACT

Glucose-stimulated insulin secretion is the hallmark of the pancreatic ß-cell, a critical player in the regulation of blood glucose concentration. In 1974, the remarkable observation was made that an efflux of intracellular inorganic phosphate (Pi) accompanied the events of stimulated insulin secretion. The mechanism behind this "phosphate flush," its association with insulin secretion, and its regulation have since then remained a mystery. We recapitulated the phosphate flush in the MIN6m9 ß-cell line and pseudoislets. We demonstrated that knockdown of XPR1, a phosphate transporter present in MIN6m9 cells and pancreatic islets, prevented this flush. Concomitantly, XPR1 silencing led to intracellular Pi accumulation and a potential impact on Ca2+ signaling. XPR1 knockdown slightly blunted first-phase glucose-stimulated insulin secretion in MIN6m9 cells, but had no significant impact on pseudoislet secretion. In keeping with other cell types, basal Pi efflux was stimulated by inositol pyrophosphates, and basal intracellular Pi accumulated following knockdown of inositol hexakisphosphate kinases. However, the glucose-driven phosphate flush occurred despite inositol pyrophosphate depletion. Finally, while it is unlikely that XPR1 directly affects exocytosis, it may protect Ca2+ signaling. Thus, we have revealed XPR1 as the missing mediator of the phosphate flush, shedding light on a 45-year-old mystery.


Subject(s)
Insulin Secretion/genetics , Insulin-Secreting Cells/metabolism , Phosphates/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Virus/metabolism , Animals , Calcium/metabolism , Exocytosis/genetics , Gene Knockdown Techniques , Glucose/metabolism , Insulin/metabolism , Islets of Langerhans/metabolism , Male , Mice , Receptors, G-Protein-Coupled/genetics , Receptors, Virus/genetics , Signal Transduction/physiology , Xenotropic and Polytropic Retrovirus Receptor
2.
Cell Signal ; 58: 131-136, 2019 06.
Article in English | MEDLINE | ID: mdl-30742888

ABSTRACT

The inositol pyrophosphate, diphosphoinositol pentakisphosphate (IP7), is thought to negatively regulate the critical insulin signaling protein Akt/PKB. Knockdown of the IP7-generating inositol hexakisphosphate kinase 1 (IP6K1) results in a concomitant increase in signaling through Akt/PKB in most cell types so far examined. Total in vivo knockout of IP6K1 is associated with a phenotype resistant to high-fat diet, due to enhanced Akt/PKB signaling in classic insulin regulated tissues, counteracting insulin resistance. In contrast, we have shown an important positive role for IP6K1 in insulin exocytosis in the pancreatic ß-cell. These cells also possess functional insulin receptors and the feedback loop following insulin secretion is a key aspect of their normal function. Thus we examined the effect of silencing IP6K1 on the activation of Akt/PKB in ß-cells. Silencing reduced the glucose-stimulated increase in Akt/PKB phosphorylation on T308 and S473. These effects were reproduced with the selective pan-IP6K inhibitor TNP. The likely explanation for IP7 reduction decreasing rather than increasing Akt/PKB phosphorylation is that IP7 is responsible for generating the insulin signal, which is the main source of Akt/PKB activation. In agreement, insulin receptor activation was compromised in TNP treated cells. To test whether the mechanism of IP7 inhibition of Akt/PKB still exists in ß-cells, we treated them at basal glucose with an insulin concentration equivalent to that reached during glucose stimulation. TNP potentiated the Akt/PKB phosphorylation of T308 induced by exogenous insulin. Thus, the IP7 regulation of ß-cell Akt/PKB is determined by two opposing forces, direct inhibition of Akt/PKB versus indirect stimulation via secreted insulin. The latter mechanism is dominant, masking the inhibitory effect. Consequently, pharmacological strategies to knock down IP6K activity might not have the same positive output in the ß-cell as in other insulin regulated tissues.


Subject(s)
Inositol Phosphates/metabolism , Insulin-Secreting Cells/metabolism , Phosphotransferases (Phosphate Group Acceptor)/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Cell Line , Insulin Secretion , Mice
4.
Cell Signal ; 46: 120-128, 2018 06.
Article in English | MEDLINE | ID: mdl-29522819

ABSTRACT

Diphosphoinositol pentakisphosphate (IP7) is critical for the exocytotic capacity of the pancreatic ß-cell, but its regulation by the primary instigator of ß-cell exocytosis, glucose, is unknown. The high Km for ATP of the IP7-generating enzymes, the inositol hexakisphosphate kinases (IP6K1 and 2) suggests that these enzymes might serve as metabolic sensors in insulin secreting ß-cells and act as translators of disrupted metabolism in diabetes. We investigated this hypothesis and now show that glucose stimulation, which increases the ATP/ADP ratio, leads to an early rise in IP7 concentration in ß-cells. RNAi mediated knock down of the IP6K1 isoform inhibits both glucose-mediated increase in IP7 and first phase insulin secretion, demonstrating that IP6K1 integrates glucose metabolism and insulin exocytosis. In diabetic mouse islets the deranged ATP/ADP levels under both basal and glucose-stimulated conditions are mirrored in both disrupted IP7 generation and insulin release. Thus the unique metabolic sensing properties of IP6K1 guarantees appropriate concentrations of IP7 and thereby both correct basal insulin secretion and intact first phase insulin release. In addition, our data suggest that a specific cell signaling defect, namely, inappropriate IP7 generation may be an essential convergence point integrating multiple metabolic defects into the commonly observed phenotype in diabetes.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Insulin-Secreting Cells/enzymology , Phosphotransferases (Phosphate Group Acceptor)/metabolism , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Animals , Diabetes Mellitus, Experimental , Gene Knockdown Techniques , Glucose/metabolism , Humans , Inositol Phosphates/metabolism , Inositol Phosphates/physiology , Insulin Secretion , Insulin-Secreting Cells/metabolism , Mice , Mice, Inbred C57BL , Phosphotransferases (Phosphate Group Acceptor)/genetics
5.
Cell Signal ; 42: 127-133, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29042286

ABSTRACT

Inositol pyrophosphates have emerged as important regulators of many critical cellular processes from vesicle trafficking and cytoskeletal rearrangement to telomere length regulation and apoptosis. We have previously demonstrated that 5-di-phosphoinositol pentakisphosphate, IP7, is at a high level in pancreatic ß-cells and is important for insulin exocytosis. To better understand IP7 regulation in ß-cells, we used an insulin secreting cell line, HIT-T15, to screen a number of different pharmacological inhibitors of inositide metabolism for their impact on cellular IP7. Although the inhibitors have diverse targets, they all perturbed IP7 levels. This made us suspicious that indirect, off-target effects of the inhibitors could be involved. It is known that IP7 levels are decreased by metabolic poisons. The fact that the inositol hexakisphosphate kinases (IP6Ks) have a high Km for ATP makes IP7 synthesis potentially vulnerable to ATP depletion. Furthermore, many kinase inhibitors are targeted to the ATP binding site of kinases, but given the similarity of such sites, high specificity is difficult to achieve. Here, we show that IP7 concentrations in HIT-T15 cells were reduced by inhibitors of PI3K (wortmannin, LY294002), PI4K (Phenylarsine Oxide, PAO), PLC (U73122) and the insulin receptor (HNMPA). Each of these inhibitors also decreased the ATP/ADP ratio. Thus reagents that compromise energy metabolism reduce IP7 indirectly. Additionally, PAO, U73122 and LY294002 also directly inhibited the activity of purified IP6K. These data are of particular concern for those studying signal transduction in pancreatic ß-cells, but also highlight the fact that employment of these inhibitors could have erroneously suggested the involvement of key signal transduction pathways in various cellular processes. Conversely, IP7's role in cellular signal transduction is likely to have been underestimated.


Subject(s)
Adenosine Triphosphate/metabolism , Enzyme Inhibitors/pharmacology , Inositol Phosphates/antagonists & inhibitors , Insulin-Secreting Cells/drug effects , Phosphotransferases (Phosphate Group Acceptor)/antagonists & inhibitors , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/antagonists & inhibitors , Androstadienes/pharmacology , Animals , Arsenicals/pharmacology , Cell Line , Chromones/pharmacology , Cricetulus , Estrenes/pharmacology , Gene Expression , Humans , Inositol Phosphates/metabolism , Insulin/biosynthesis , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Morpholines/pharmacology , Phosphotransferases (Phosphate Group Acceptor)/genetics , Phosphotransferases (Phosphate Group Acceptor)/metabolism , Pyrrolidinones/pharmacology , Receptor, Insulin/pharmacology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Succinimides/pharmacology , Triazoles/pharmacology , Wortmannin
6.
Integr Biol (Camb) ; 4(2): 209-19, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22267247

ABSTRACT

Pancreatic alpha cells contribute to glucose homeostasis by the regulated secretion of glucagon, which increases glycogenolysis and hepatic gluconeogenesis in response to hypoglycemia. Alterations of glucagon secretion are observed in diabetic patients and exacerbate the disease. The restricted availability of purified primary alpha cells has limited our understanding of their function in health and disease. This study was designed to establish convenient protocols for the purification of viable alpha cells from rat and human pancreatic islets by FACS, using intrinsic cellular properties. Islets were isolated from the pancreata of Wistar rats or deceased human organ donors. Dispersed islet cells were separated by FACS based on light scatter and autofluorescence. Purity of sorted cells was evaluated by immunocytochemistry using hormone specific antibodies. Relative hormone expression was further determined by quantitative RT-PCR. Viability was determined by Annexin V and propidium iodide staining and function was assessed by monitoring cytoplasmic free Ca(2+) concentration ([Ca(2+)](i)) using Fura-2/AM. We developed species-specific FACS gating strategies that resulted in populations consisting mainly of alpha cells (96.6 ± 1.4%, n = 3 for rat; 95.4 ± 1.7%, n = 4 for human, mean ± SEM). These cell fractions showed ~5-fold and ~4-fold enrichment (rat and human, respectively) of glucagon mRNA expression compared to total ungated islet cells. Most of the sorted cells were viable and functional, as they responded with an increase in [Ca(2+)](i) upon stimulation with L-arginine (10 mM). The majority of the sorted human alpha cells responded also to stimulation with kainate (100 µM), whereas this response was infrequent in rat alpha cells. Using the same sample preparation, but a different gating strategy, we were also able to sort rat and human populations enriched in beta cells. In conclusion, we have simplified and optimized a method for the purification of rat alpha cells, as well as established a novel approach to separate human alpha cells using neither antibodies nor dyes possibly interfering with cellular functions.


Subject(s)
Flow Cytometry/methods , Glucagon-Secreting Cells/cytology , Islets of Langerhans/cytology , Adult , Aged , Animals , Calcium/analysis , Cell Survival , Female , Glucagon-Secreting Cells/chemistry , Humans , Islets of Langerhans/chemistry , Male , Middle Aged , RNA/chemistry , RNA/genetics , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...