Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










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.
Sci Rep ; 10(1): 20145, 2020 11 19.
Article in English | MEDLINE | ID: mdl-33214580

ABSTRACT

The secretion of glucagon by pancreatic alpha cells is regulated by a number of external and intrinsic factors. While the electrophysiological processes linking a lowering of glucose concentrations to an increased glucagon release are well characterized, the evidence for the identity and function of the glucose sensor is still incomplete. In the present study we aimed to address two unsolved problems: (1) do individual alpha cells have the intrinsic capability to regulate glucagon secretion by glucose, and (2) is glucokinase the alpha cell glucose sensor in this scenario. Single cell RT-PCR was used to confirm that glucokinase is the main glucose-phosphorylating enzyme expressed in rat pancreatic alpha cells. Modulation of glucokinase activity by pharmacological activators and inhibitors led to a lowering or an increase of the glucose threshold of glucagon release from single alpha cells, measured by TIRF microscopy, respectively. Knockdown of glucokinase expression resulted in a loss of glucose control of glucagon secretion. Taken together this study provides evidence for a crucial role of glucokinase in intrinsic glucose regulation of glucagon release in rat alpha cells.


Subject(s)
Glucagon-Secreting Cells/metabolism , Glucagon/metabolism , Glucokinase/metabolism , Glucose/metabolism , Animals , Biosensing Techniques , Fluorescent Antibody Technique , Gene Expression Regulation, Enzymologic , Glucagon/genetics , Glucagon-Secreting Cells/drug effects , Glucokinase/genetics , Glucose/pharmacology , Isoenzymes/metabolism , Mannoheptulose/pharmacology , Microscopy, Fluorescence , Rats, Wistar , Single-Cell Analysis/methods , Sulfones/pharmacology , Thiazoles/pharmacology
3.
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
5.
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
6.
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
7.
Proc Natl Acad Sci U S A ; 112(20): E2611-9, 2015 May 19.
Article in English | MEDLINE | ID: mdl-25941406

ABSTRACT

Insulin resistance and ß-cell failure are the major defects in type 2 diabetes mellitus. However, the molecular mechanisms linking these two defects remain unknown. Elevated levels of apolipoprotein CIII (apoCIII) are associated not only with insulin resistance but also with cardiovascular disorders and inflammation. We now demonstrate that local apoCIII production is connected to pancreatic islet insulin resistance and ß-cell failure. An increase in islet apoCIII causes promotion of a local inflammatory milieu, increased mitochondrial metabolism, deranged regulation of ß-cell cytoplasmic free Ca(2+) concentration ([Ca(2+)]i) and apoptosis. Decreasing apoCIII in vivo results in improved glucose tolerance, and pancreatic apoCIII knockout islets transplanted into diabetic mice, with high systemic levels of the apolipoprotein, demonstrate a normal [Ca(2+)]i response pattern and no hallmarks of inflammation. Hence, under conditions of islet insulin resistance, locally produced apoCIII is an important diabetogenic factor involved in impairment of ß-cell function and may thus constitute a novel target for the treatment of type 2 diabetes mellitus.


Subject(s)
Apolipoprotein C-III/metabolism , Diabetes Mellitus, Type 2/physiopathology , Insulin Resistance/physiology , Insulin-Secreting Cells/pathology , Analysis of Variance , Animals , Apolipoprotein C-III/genetics , Blotting, Western , Calcium/metabolism , Cell Line, Tumor , Immunohistochemistry , Mice , Mice, Knockout , Microscopy, Confocal , Mitochondria/metabolism , Real-Time Polymerase Chain Reaction
8.
Exp Cell Res ; 336(1): 158-70, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-25983130

ABSTRACT

Strategies for correct diagnosis, treatment evaluation and recurrence prediction are important for the prognosis and mortality rates among cancer patients. In spite of major improvements in clinical management, gastrointestinal stromal tumors (GISTs) can still be deadly due to metastasis and recurrences, which confirms the unmet need of reliable follow-up modalities. Tumor-specific secreted, shed or leaked proteins (collectively known as secretome) are considered promising sources for biomarkers, and suitable for detection in biofluids. Herein, we stimulated cell secretion in the imatinib-sensitive GIST882 cell line and profiled the secretome, collected as conditioned media, by using a shotgun proteomics approach. We identified 764 proteins from all conditions combined, 51.3% being predicted as classically/non-classically secreted. The protein subsets found were dependent on the stimulatory condition. The significant increase in protein release by the classical pathway was strongly associated with markers already found in other cancer types. Furthermore, most of the released proteins were non-classically released and overlapped to a high degree with proteins of exosomal origin. Imatinib pre-treatment radically changed these secretory patterns, which can have clinical implications when investigating biomarkers in imatinib-treated versus non-treated GIST patients. Our results show, for the first time, that GISTs contain a secretome signature. In the search for suitable biomarkers in the more complex GIST patient samples, this study aids in the understanding of basic GIST secretome characteristics.


Subject(s)
Gastrointestinal Stromal Tumors/metabolism , Insulin-Secreting Cells/metabolism , Neoplasm Proteins/metabolism , Proteome/metabolism , Proteomics/methods , Animals , Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Blotting, Western , Cells, Cultured , Chromatography, Liquid/methods , Gastrointestinal Stromal Tumors/drug therapy , Gastrointestinal Stromal Tumors/pathology , Humans , Imatinib Mesylate , Insulin-Secreting Cells/cytology , Mice , Piperazines/pharmacology , Pyrimidines/pharmacology , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods
9.
Exp Cell Res ; 319(8): 1229-38, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23499741

ABSTRACT

Gastrointestinal stromal tumors (GISTs) are thought to originate from the electrically active pacemaker cells of the gastrointestinal tract. Despite the presence of synaptic-like vesicles and proteins involved in cell secretion it remains unclear whether GIST cells possess regulated release mechanisms. The GIST tumor cell line GIST882 was used as a model cell system, and stimulus-release coupling was investigated by confocal microscopy of cytoplasmic free Ca(2+) concentration ([Ca(2+)]i), flow cytometry, and luminometric measurements of extracellular ATP. We demonstrate that GIST cells have an intact intracellular Ca(2+)-signaling pathway that regulates ATP release. Cell viability and cell membrane integrity was preserved, excluding ATP leakage due to cell death and suggesting active ATP release. The stimulus-secretion signal transduction is at least partly dependent on Ca(2+) influx since exclusion of extracellular Ca(2+) diminishes the ATP release. We conclude that measurements of ATP release in GISTs may be a useful tool for dissecting the signal transduction pathway, mapping exocytotic components, and possibly for the development and evaluation of drugs. Additionally, release of ATP from GISTs may have importance for tumor tissue homeostasis and immune surveillance escape.


Subject(s)
Adenosine Triphosphate/metabolism , Calcium/pharmacology , Gastrointestinal Neoplasms/metabolism , Gastrointestinal Stromal Tumors/metabolism , Animals , Cations/pharmacology , Cell Line, Tumor , Cell Membrane Permeability/drug effects , DNA Mutational Analysis , Gastrointestinal Neoplasms/genetics , Gastrointestinal Neoplasms/pathology , Gastrointestinal Stromal Tumors/genetics , Gastrointestinal Stromal Tumors/pathology , HEK293 Cells , Humans , Insulin/metabolism , Mice , Phenotype , Proto-Oncogene Proteins c-kit/genetics
10.
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
11.
Chem Biol Interact ; 188(2): 301-8, 2010 Nov 05.
Article in English | MEDLINE | ID: mdl-20399200

ABSTRACT

Methylmercury is a widely distributed environmental toxicant with detrimental effects on the developing and adult nervous system. Due to its accumulation in the food chain, chronic exposure to methylmercury via consumption of fish and sea mammals is still a major concern for human health, especially developmental exposure that may lead to neurological alterations, including cognitive and motor dysfunctions. Mercury-induced neurotoxicity and the identification of the underlying mechanisms has been a main focus of research in the neurotoxicology field. Three major mechanisms have been identified as critical in methylmercury-induced cell damage including (i) disruption of calcium homeostasis, (ii) induction of oxidative stress via overproduction of reactive oxygen species or reduction of antioxidative defenses and (iii) interactions with sulfhydryl groups. In vivo and in vitro studies have provided solid evidence for the occurrence of neural cell death, as well as cytoarchitectural alterations in the nervous system after exposure to methylmercury. Signaling cascades leading to cell death induced by methylmercury involve the release of mitochondrial factors, such as cytochrome c and AIF with subsequent caspase-dependent or -independent apoptosis, respectively; induction of calcium-dependent proteases calpains; interaction with lysosomes leading to release of cathepsins. Interestingly, several pathways can be activated in parallel, depending on the cell type. In this paper, we provide an overview of recent findings on methylmercury-induced neurotoxicity and cell death pathways that have been described in neural and endocrine cell systems.


Subject(s)
Methylmercury Compounds/toxicity , Animals , Apoptosis , Endocrine System/drug effects , Humans , Methylmercury Compounds/metabolism , Nervous System/drug effects , Neurons/cytology , Neurons/drug effects , Neurons/metabolism
12.
Physiol Behav ; 95(5): 668-76, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-18930070

ABSTRACT

We have examined behavioral consequences of genetic deletion of the adenosine A3 receptors in mice. The open field behavior of A3 adenosine receptor knock-out (A3R KO) mice was investigated both under basal conditions and after stimulation with psychostimulants. Adolescent (21 day-old) and adult A3R KO males showed an increase in overall motor activity compared to wild type (WT) males, but the type of activity differed. The motor activity, especially rearing, was also higher in A3R KO compared to WT adult females. A3 receptors have a low affinity for caffeine and it was therefore surprising to find a decreased response to stimulation with either caffeine or amphetamine in A3R KO as compared to WT mice in males as well as females. Telemetry recordings also showed a significantly smaller increase in activity upon darkness in A3R KO. There were no compensatory changes in the mRNA expression of any other adenosine receptor subtypes (A1, A2A and A2B) or any changes in dopamine D1 and D2 receptor binding in A3R KO brains. Challenge with the developmental toxicant methylmercury (1 microM in drinking water) during pregnancy and lactation did not cause any behavioral alterations in adolescent and adult WT female offspring. In contrast, the A3R KO female offspring displayed changes in locomotion indicating an interaction between perinatal methylmercury and adenosine A3 receptors. In conclusion, despite low expression of A3 receptors in wild type mouse brain we observed several behavioral consequences of genetic elimination of the adenosine A3 receptors. The possibility that this is due to a role of A3 receptors in development is discussed.


Subject(s)
Caffeine/pharmacology , Central Nervous System Stimulants/pharmacology , Exploratory Behavior/physiology , Motor Activity/drug effects , Receptor, Adenosine A3/physiology , Amphetamine/pharmacology , Analysis of Variance , Animals , Darkness , Environmental Pollutants/toxicity , Exploratory Behavior/drug effects , Female , Male , Methylmercury Compounds/toxicity , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/genetics , Neurotoxins/toxicity , Pregnancy , Prenatal Exposure Delayed Effects , Receptor, Adenosine A3/drug effects , Receptor, Adenosine A3/genetics , Receptors, Dopamine/classification , Receptors, Dopamine/drug effects , Receptors, Dopamine/metabolism , Sex Factors , Statistics, Nonparametric
13.
Eur J Pharmacol ; 596(1-3): 6-13, 2008 Oct 31.
Article in English | MEDLINE | ID: mdl-18727925

ABSTRACT

Brain levels of adenosine are elevated during hypoxia. Through effects on adenosine receptors (A(1), A(2A), A(2B) and A(3)) on astrocytes, adenosine can influence functions such as glutamate uptake, reactive gliosis, swelling, as well as release of neurotrophic and neurotoxic factors having an impact on the outcome of metabolic stress. We have studied the roles of these receptors in astrocytes by evaluating their susceptibility to damage induced by oxygen deprivation or exposure to the hypoxia mimic cobalt chloride (CoCl(2)). Hypoxia caused ATP breakdown and purine release, whereas CoCl(2) (0.8 mM) mainly reduced ATP by causing cell death in human D384 astrocytoma cells. Further experiments were conducted in primary astrocytes prepared from specific adenosine receptor knock-out (KO) and wild type (WT) mice. In WT cells purine release following CoCl(2) exposure was mainly due to nucleotide release, whereas hypoxia-induced intracellular ATP breakdown followed by nucleoside efflux. N-ethylcarboxamidoadenosine (NECA), an unselective adenosine receptor agonist, protected from cell death following hypoxia. Cytotoxicity was more pronounced in A(1)R KO astrocytes and tended to be higher in WT cells in the presence of the A(1) receptor antagonist 8-cyclopentyl-1,3-dipropylxanthine (DPCPX). Genetic deletion of A(2A) receptor resulted in less prominent effects. A(3)R KO glial cells were more affected by hypoxia than WT cells. Accordingly, the A(3) receptor agonist 2-chloro-N(6)-(3-iodobenzyl)-N-methyl-5'-carbamoyladenosine (CL-IB-MECA) reduced ATP depletion caused by hypoxic conditions. It also reduced apoptosis in human astroglioma D384 cells after oxygen deprivation. In conclusion, the data point to a cytoprotective role of adenosine mediated by both A(1) and A(3) receptors in primary mouse astrocytes.


Subject(s)
Astrocytes/metabolism , Receptor, Adenosine A1/physiology , Receptor, Adenosine A3/physiology , Adenosine/metabolism , Adenosine A1 Receptor Agonists , Adenosine A3 Receptor Agonists , Adenosine Triphosphate/metabolism , Animals , Animals, Newborn , Apoptosis , Astrocytes/cytology , Cell Hypoxia , Cell Survival , Cells, Cultured , Cobalt/pharmacology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Mice , Mice, Knockout , Purines/metabolism , Receptor, Adenosine A1/genetics , Receptor, Adenosine A2A/genetics , Receptor, Adenosine A2A/physiology , Receptor, Adenosine A3/genetics
14.
Toxicology ; 241(3): 119-33, 2007 Nov 30.
Article in English | MEDLINE | ID: mdl-17920182

ABSTRACT

Adenosine and its receptors are, as part of the brain stress response, potential targets for neuroprotective drugs. We have investigated if the adenosine receptor system affects the developmental neurotoxicity caused by the fish pollutant methylmercury (MeHg). Behavioral outcomes of low dose perinatal MeHg exposure were studied in mice where the A(1) and A(2A) adenosine receptors were either partially blocked by caffeine treatment or eliminated by genetic modification (A(1)R and A(2A)R knock-out mice). From gestational day 7 to day 7 of lactation dams were administered doses that mimic human intake via normal diet, i.e. 1microM MeHg and/or 0.3g/l caffeine in the drinking water. This exposure to MeHg resulted in a doubling of brain Hg levels in wild type females and males at postnatal day 21 (PND21). Open field analysis was performed at PND21 and 2 months of age. MeHg caused time-dependent behavioral alterations preferentially in male mice. A decreased response to amphetamine in 2-month-old males pointed to disturbances in dopaminergic functions. Maternal caffeine intake induced long-lasting changes in the offspring evidenced by an increased motor activity and a modified response to psychostimulants in adult age, irrespectively of sex. Similar alterations were observed in A(1)R knock-out mice, suggesting that adenosine A(1) receptors are involved in the alterations triggered by caffeine exposure during development. Perinatal caffeine treatment and, to some extent, genetic elimination of adenosine A(1) receptors, attenuated the behavioral consequences of MeHg in males. Importantly, also deletion of the A(2A) adenosine receptor reduced the vulnerability to MeHg, consistent with the neuroprotective effects of adenosine A(2A) receptor inactivation observed in hypoxia and Parkinson's disease. Thus, the consequences of MeHg toxicity during gestation and lactation can be reduced by adenosine A(1) and A(2A) receptor inactivation, either via their genetic deletion or by treatment with their antagonist caffeine.


Subject(s)
Aging , Caffeine/pharmacology , Environmental Pollutants/toxicity , Gene Deletion , Methylmercury Compounds/toxicity , Motor Activity/drug effects , Receptors, Purinergic P1/genetics , Aging/drug effects , Aging/genetics , Aging/metabolism , Animals , Brain/drug effects , Brain/growth & development , Brain/metabolism , Environmental Pollutants/pharmacokinetics , Female , Lactation , Male , Maternal Exposure/adverse effects , Methylmercury Compounds/pharmacokinetics , Mice , Mice, Knockout , Motor Activity/genetics , Pregnancy , Purinergic P1 Receptor Antagonists , Sex Factors
15.
Physiol Behav ; 92(1-2): 15-20, 2007 Sep 10.
Article in English | MEDLINE | ID: mdl-17574632

ABSTRACT

Adenosine is an endogenous neuromodulator, acting on four distinctive G-protein-coupled receptors, the A1, A2A, A2B and A3 adenosine receptors. Increased neuronal activity and, hypoxia or ischemia, result in elevated levels of adenosine reflecting changes of the metabolic state. This increases activation of the adenosine receptors. It is well appreciated that adenosine has a neuroprotective role in brain injuries. Although adenosine effects have been explained mainly by actions on nerve cells, modulation of glial functions by adenosine is likely to be important as discussed in this minireview. Thus, in astrocytes adenosine receptors modulate inter alia glycogen metabolism, glutamate transporters, astrogliosis and astrocyte swelling. Microglial cells appear to be important in regulating adenosine formation from ATP and adenosine can affect many microglial signaling pathways. Adenosine receptors on oligodendrocytes regulate white matter development.


Subject(s)
Adenosine/metabolism , Neuroglia/metabolism , Receptors, Purinergic P1/metabolism , Animals , Humans , Neuroglia/cytology
16.
Biochem Pharmacol ; 73(5): 694-708, 2007 Mar 01.
Article in English | MEDLINE | ID: mdl-17169330

ABSTRACT

Cobalt is suspected to cause memory deficit in humans and was reported to induce neurotoxicity in animal models. We have studied the effects of cobalt in primary cultures of mouse astrocytes. CoCl(2) (0.2-0.8mM) caused dose-dependent ATP depletion, apoptosis (cell shrinkage, phosphatidylserine externalization and chromatin rearrangements) and secondary necrosis. The mitochondria appeared to be a main target of cobalt toxicity, as shown by the loss of mitochondrial membrane potential (DeltaPsi(m)) and release from the mitochondria of apoptogenic factors, e.g. apoptosis inducing factor (AIF). Pre-treatment with bongkrekic acid reduced ATP depletion, implicating the involvement of the mitochondrial permeability transition (MPT) pore. Cobalt increased the generation of oxygen radicals, but antioxidants did not prevent toxicity. There was also an impaired response to ATP stimulation, evaluated as a lower raise in intracellular calcium. Similarly to hypoxia and dymethyloxallyl glycine (DMOG), cobalt triggered stabilization of the alpha-subunit of hypoxia-inducible factor HIF-1 (HIF-1alpha). This early event was followed by an increased expression of HIF-1 regulated genes, e.g. stress protein HO-1, pro-apoptotic factor Nip3 and iNOS. Although all of the three stimuli activated the HIF-1alpha pathway and decreased ATP levels, the downstream effects were different. DMOG only inhibited cell proliferation, whereas the other two conditions caused cell death by apoptosis and necrosis. This points to cobalt and hypoxia not only inducing HIF-1alpha regulated genes but also affecting similarly other cellular functions, including metabolism.


Subject(s)
Astrocytes/drug effects , Cell Hypoxia , Cobalt/toxicity , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Oxygen/metabolism , Amino Acids, Dicarboxylic/pharmacology , Animals , Calcium Signaling , Cells, Cultured , Dose-Response Relationship, Drug , Mice , Reactive Oxygen Species
17.
Am J Physiol Gastrointest Liver Physiol ; 288(5): G972-7, 2005 May.
Article in English | MEDLINE | ID: mdl-15637180

ABSTRACT

Adenosine is known to stimulate chloride secretion by mouse jejunum. Whereas the receptor on the basolateral side is believed to be A2B, the receptor involved in the luminal effect of adenosine has not been identified. We found that jejuna expressed mRNA for all adenosine receptor subtypes. In this study, we investigated the stimulation of chloride secretion by adenosine in jejuna derived from mice lacking the adenosine receptors of A1 (A1R) and A2A (A(2A)R) or control littermates. The jejunal epithelium was mounted in a Ussing chamber, and a new method on the basis of impedance analysis was used to calculate the short-circuit current (I(sc)) values. Chloride secretion was assessed by the I(sc) after inhibition of the sodium-glucose cotransporter by adding phloridzin to the apical bathing solution. The effect of apical adenosine on chloride secretion was lost in jejuna from mice lacking the A1R. There was no difference in the response to basolaterally applied adenosine or to apical forskolin. Furthermore, in jejuna from control mice, the effect of apical adenosine was also abolished in the presence of 8-cyclopentyl-1,3-dipropylxanthine, a specific A1R antagonist. Responses to adenosine were identical in jejuna from control and A(2A)R knockout mice. This study demonstrates that A1R (and not A(2A)R) mediates the enhancement of chloride secretion induced by luminal adenosine in mice jejunum.


Subject(s)
Adenosine/physiology , Chlorides/physiology , Jejunum/physiology , Receptor, Adenosine A1/physiology , Receptors, Adenosine A2/physiology , Animals , Gene Expression/physiology , Jejunum/metabolism , Mice , Mice, Knockout , Receptor, Adenosine A1/genetics , Receptors, Adenosine A2/genetics
18.
Naunyn Schmiedebergs Arch Pharmacol ; 370(4): 270-8, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15378230

ABSTRACT

The adenosine receptor agonist 2-[ p-(2-carboxyethyl)phenylethylamino]-5'- N-ethylcarboxamidoadenosine (CGS 21680) is generally considered to be a selective adenosine A(2A) receptor ligand. However, the compound has previously been shown to exhibit binding characteristics that are not compatible with adenosine A(2A) receptor binding, at least in brain regions other than the striatum. We have examined binding of [(3)H]CGS 21680 and of antagonist radioligands with high selectivity for adenosine A(1) or A(2A) receptors to hippocampus and striatum of mice lacking either adenosine A(1) (A1R((-/-))) or A(2A) (A2AR((-/-))) receptors. Both receptor autoradiography and membrane binding techniques were used for this purpose and gave similar results. There were no significant changes in the binding of the A(1) receptor antagonist [(3)H]DPCPX in mice lacking A(2A) receptors, or in the binding of the A(2A) receptor antagonists [(3)H]SCH 58261 and [(3)H]ZM 241385 in mice lacking A(1) receptors. Furthermore, [(3)H]CGS 21680 binding in striatum was abolished in the A2AR((-/-)), and essentially unaffected in striatum from mice lacking A(1) receptors. In hippocampus, however, binding of [(3)H]CGS 21680 remained in the A2AR((-/-)), whereas binding was virtually abolished in the A1R((-/-)). There were no adaptive alterations in A(2A) receptor expression in this region in A1R((-/-)) mice. Thus, most of the [(3)H]CGS 21680 binding in hippocampus is dependent on the presence of adenosine A(1) receptors, but not on A(2A) receptors, indicating a novel binding site or novel binding mode.


Subject(s)
Adenosine/analogs & derivatives , Adenosine/metabolism , Hippocampus/metabolism , Phenethylamines/metabolism , Receptor, Adenosine A1/metabolism , Animals , Ligands , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Binding/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Adenosine A1/deficiency , Receptor, Adenosine A1/genetics
19.
Naunyn Schmiedebergs Arch Pharmacol ; 367(5): 500-8, 2003 May.
Article in English | MEDLINE | ID: mdl-12684742

ABSTRACT

In the present study we have investigated the neurotoxic effects of the exposure to a low dose (0.5 mg/kg/day) of methylmercury (MeHg) on the developing nervous system. Pregnant rats were treated with MeHg from day 7 of pregnancy to day 7 of lactation. At postnatal day 20 the offspring did not display prominent functional cerebellar alterations, as evaluated by the Rotarod performance. Motor activity (locomotion, rearing and motility) was tested in the 21-day-old rats after administration of apomorphine, an agonist of D(1), D(2), and D(3) dopamine receptors. A low dose of apomorphine (0.1 mg/kg) induced a significantly stronger increase in motility and locomotion in MeHg-treated rats as compared to controls. The same effect was also observed in rats injected with 1 mg/kg apomorphine. No changes were observed in rearing at either doses of the dopamine receptor agonist. The data suggest that changes in dopaminergic transmission are induced by exposure to MeHg in early life. The expression of the striatal dopamine D(1) and D(2) receptors was examined by in situ hybridization in the striatum of the 21-day-old rats. The analysis did not reveal any significant changes at the mRNA level. Ligand autoradiography experiments showed a significant reduction in dopamine D(2) receptor binding in the caudate putamen of MeHg-treated rats. Spatial learning ability was tested in 2-month-old rats using the Morris swim maze test. Changes in retention were shown in MeHg-treated rats, indicating that MeHg induced memory alterations. Taken together, these findings show that exposure to a very low dose of MeHg during development exerts neurotoxic effects on the dopaminergic system and that alterations of brain functions persist in adult life.


Subject(s)
Central Nervous System/metabolism , Methylmercury Compounds/toxicity , Motor Activity/drug effects , Prenatal Exposure Delayed Effects , Receptors, Dopamine D1/drug effects , Receptors, Dopamine D2/drug effects , Animals , Animals, Newborn/physiology , Apomorphine/administration & dosage , Apomorphine/pharmacokinetics , Central Nervous System/growth & development , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Dopamine/physiology , Female , In Situ Hybridization , Maze Learning/drug effects , Methylmercury Compounds/administration & dosage , Motor Activity/physiology , Pregnancy , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Dopamine D1/genetics , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D2/genetics , Receptors, Dopamine D2/metabolism , Rotarod Performance Test , Swimming
20.
Am J Nephrol ; 23(3): 140-51, 2003.
Article in English | MEDLINE | ID: mdl-12624487

ABSTRACT

BACKGROUND: Pyelonephritis is a risk factor for renal tubular epithelial cell damage. Recent studies have shown that Escherichia coli and/or its toxins may stimulate apoptotic cell death in renal tubular cells, but the underlying molecular mechanisms remain to be elucidated. METHODS: Confluent LLC-PK(1) cells were exposed to E. coli toxins from overnight cultures of the uropathogenic O6K13H1 (O6) and the nonpathogenic W3110. The cell death was studied with morphological and biological assay. RESULTS: E. coli soluble toxins from uropathogenic O6:K13:H1(O6) strain were found to induce apoptosis in a dose- and time-dependent manner in LLC-PK1 cells. The expression of FasR and the phosphorylation of ERK1/2 were significantly upregulated by O6 soluble toxins in a time-dependent manner. Cell death was completely inhibited by two specific ERK1/2 inhibitors, but not by a broad caspase inhibitor, zVAD-fmk, implicating a caspase-independent pathway via ERK. Moreover, we found that lysophosphatidic acid could trigger a survival signal through G-proteins and PI3K. CONCLUSION: We demonstrate that apoptosis induced by uropathogenic E. coli toxins is dependent on ERK1/2. Caspases, although being activated, are not necessary for cell death, and they act after the ERK signaling at which point cells become committed to cell death or can be rescued.


Subject(s)
Apoptosis/drug effects , Bacterial Toxins/pharmacology , Caspases/physiology , Kidney Tubules, Proximal/drug effects , Mitogen-Activated Protein Kinases/physiology , Pyelonephritis/pathology , Analysis of Variance , Animals , Blotting, Western , Cells, Cultured , Escherichia coli , Kidney Tubules, Proximal/enzymology , Pyelonephritis/enzymology , Signal Transduction , Swine , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...