Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Mol Cell Endocrinol ; 407: 18-25, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25766501

ABSTRACT

INTRODUCTION: Autosomal dominant hypocalcaemia (ADH) is caused by activating mutations in the calcium sensing receptor gene (CaR) and characterised by mostly asymptomatic mild to moderate hypocalcaemia with low, inappropriately serum concentration of PTH. OBJECTIVE: The purpose of the present study was to biochemically and functionally characterise a novel mutation of CaR. PATIENTS: A female proband presenting with hypocalcaemia was diagnosed to have "idiopathic hypoparathyroidism" at the age of 10 with a history of muscle pain and cramps. Further examinations demonstrated hypocalcaemia in nine additional family members, affecting three generations. MAIN OUTCOME MEASURE: P136L CaR mutation was predicted to cause gain of function of CaR. RESULTS: Affected family members showed relevant hypocalcaemia (mean ± SD; 1.9 ± 0.1 mmol/l). Patient history included mild seizures and recurrent nephrolithiasis. Genetic analysis confirmed that hypocalcaemia cosegregated with a heterozygous mutation at codon 136 (CCC → CTC/Pro → Leu) in exon 3 of CaR confirming the diagnosis of ADH. For in vitro studies P136L mutant CaR was generated by site-directed mutagenesis and examined in transiently transfected HEK293 cells. Extracellular calcium stimulation of transiently transfected HEK293 cells showed significantly increased intracellular Ca(2+) mobilisation and MAPK activity for mutant P136L CaR compared to wild type CaR. CONCLUSIONS: The present study gives insight about a novel activating mutation of CaR and confirms that the novel P136L-CaR mutation is responsible for ADH in this family.


Subject(s)
Hypercalciuria/genetics , Hypocalcemia/genetics , Hypoparathyroidism/congenital , Hypoparathyroidism/genetics , Mutation , Parathyroid Hormone/genetics , Receptors, Calcium-Sensing/genetics , Adult , Calcium/metabolism , Child , Child, Preschool , Codon , Exons , Female , Gene Expression , HEK293 Cells , Heterozygote , Humans , Hypercalciuria/complications , Hypercalciuria/metabolism , Hypercalciuria/pathology , Hypocalcemia/complications , Hypocalcemia/metabolism , Hypocalcemia/pathology , Hypoparathyroidism/complications , Hypoparathyroidism/metabolism , Hypoparathyroidism/pathology , Infant , Male , Middle Aged , Parathyroid Hormone/deficiency , Pedigree , Transfection
2.
J Lipid Res ; 56(1): 60-9, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25385827

ABSTRACT

Sphingosine 1-phosphate (S1P) is an extra- and intracellular mediator that regulates cell growth, survival, migration, and adhesion in many cell types. S1P lyase is the enzyme that irreversibly cleaves S1P and thereby constitutes the ultimate step in sphingolipid catabolism. It has been reported previously that embryonic fibroblasts from S1P lyase-deficient mice (Sgpl1(-/-)-MEFs) are resistant to chemotherapy-induced apoptosis through upregulation of B cell lymphoma 2 (Bcl-2) and Bcl-2-like 1 (Bcl-xL). Here, we demonstrate that the transporter proteins Abcc1/MRP1, Abcb1/MDR1, Abca1, and spinster-2 are upregulated in Sgpl1(-/-)-MEFs. Furthermore, the cells efficiently sequestered the substrates of Abcc1 and Abcb1, fluo-4 and doxorubicin, in subcellular compartments. In line with this, Abcb1 was localized mainly at intracellular vesicular structures. After 16 h of incubation, wild-type MEFs had small apoptotic nuclei containing doxorubicin, whereas the nuclei of Sgpl1(-/-)-MEFs appeared unchanged and free of doxorubicin. A combined treatment with the inhibitors of Abcb1 and Abcc1, zosuquidar and MK571, respectively, reversed the compartmentalization of doxorubicin and rendered the cells sensitive to doxorubicin-induced apoptosis. It is concluded that upregulation of multidrug resistance transporters contributes to the chemoresistance of S1P lyase-deficient MEFs.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Aldehyde-Lyases/deficiency , Drug Resistance, Neoplasm , Fibroblasts/drug effects , Up-Regulation/drug effects , Aniline Compounds/pharmacology , Animals , Doxorubicin/pharmacology , Fibroblasts/cytology , Fibroblasts/enzymology , Intracellular Space/drug effects , Intracellular Space/metabolism , Mice , Protein Transport/drug effects , Xanthenes/pharmacology
3.
Biochem Pharmacol ; 81(5): 617-25, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21163254

ABSTRACT

Sphingosine-1-phosphate (S1P) acts as high affinity agonist at specific G-protein-coupled receptors, S1P(1-5), that play important roles e.g. in the cardiovascular and immune systems. A S1P receptor modulating drug, FTY720 (fingolimod), has been effective in phase III clinical trials for multiple sclerosis. FTY720 is a sphingosine analogue and prodrug of FTY720-phosphate, which activates all S1P receptors except S1P(2) and disrupts lymphocyte trafficking by internalizing the S1P(1) receptor. Cis-4-methylsphingosine (cis-4M-Sph) is another synthetic sphingosine analogue that is readily taken up by cells and phosphorylated to cis-4-methylsphingosine-1-phosphate (cis-4M-S1P). Therefore, we analysed whether cis-4M-Sph interacted with S1P receptors through its metabolite cis-4M-S1P in a manner similar to FTY720. Indeed, cis-4M-Sph caused an internalization of S1P receptors, but differed from FTY720 as it acted on S1P(2) and S1P(3) and only weakly on S1P(1), while FTY720 internalized S1P(1) and S1P(3) but not S1P(2). Consequently, pre-incubation with cis-4M-Sph specifically desensitized S1P-induced [Ca(2+)](i) increases, which are mediated by S1P(2) and S1P(3), in a time- and concentration-dependent manner. This effect was not shared by sphingosine or FTY720, indicating that metabolic stability and targeting of S1P(2) receptors were important. The desensitization of S1P-induced [Ca(2+)](i) increases was dependent on the expression of SphKs, predominantly of SphK2, and thus mediated by cis-4M-S1P. In agreement, cis-4M-S1P was detected in the supernatants of cells exposed to cis-4M-Sph. It is concluded that cis-4M-Sph, through its metabolite cis-4M-S1P, acts as a S1P receptor modulator and causes S1P receptor internalization and desensitization. The data furthermore help to define requirements for sphingosine kinase substrates as S1P receptor modulating prodrugs.


Subject(s)
Receptors, Lysosphingolipid/agonists , Sphingosine/analogs & derivatives , Calcium/metabolism , Fingolimod Hydrochloride , HEK293 Cells , Humans , Lysophospholipids/pharmacology , Phosphorylation , Phosphotransferases (Alcohol Group Acceptor)/biosynthesis , Propylene Glycols/pharmacology , Sphingosine/metabolism , Sphingosine/pharmacology , Stereoisomerism
4.
Circulation ; 122(22): 2313-22, 2010 Nov 30.
Article in English | MEDLINE | ID: mdl-21098434

ABSTRACT

BACKGROUND: Hyaluronan is thought to mediate neointimal hyperplasia but also vasoprotection as an integral component of the endothelial glycocalyx. The present study addressed for the first time the effects of long-term pharmacological inhibition of hyaluronan synthesis on vascular function and atherosclerosis. METHODS AND RESULTS: Four-week-old apolipoprotein E-deficient mice on a Western diet received orally an inhibitor of hyaluronan synthesis, 4-methylumbelliferone (4-MU; 10 mg/g body wt), resulting in 600 nmol/L 4-MU in plasma. As a result, aortic plaque burden was markedly increased at 25 weeks. Furthermore, acetylcholine-dependent relaxation of aortic rings was decreased and mean arterial blood pressure was increased in response to 4-MU. However, hydralazine blunted the hypertensive effect of 4-MU without inhibiting the proatherosclerotic effect. A photothrombosis model revealed a prothrombotic state that was not due to increased platelet activation or increased thrombin activation as monitored by CD62P expression and the endogenous thrombin potential. Importantly, increased recruitment of macrophages to vascular lesions was detected after 2 and 21 weeks of 4-MU treatment by immunohistochemistry, by intravital microscopy, and in a peritonitis model. As a potential underlying mechanism, severe damage of the endothelial glycocalyx after 2 and 21 weeks of treatment with 4-MU was detected by electron microscopy of the innominate artery and myocardial capillaries. Furthermore, 600 nmol/L 4-MU inhibited hyaluronan synthesis in cultured endothelial cells. CONCLUSIONS: The data suggest that systemic inhibition of hyaluronan synthesis by 4-MU interferes with the protective function of the endothelial glycocalyx, thereby facilitating leukocyte adhesion, subsequent inflammation, and progression of atherosclerosis.


Subject(s)
Atherosclerosis/metabolism , Disease Progression , Hyaluronic Acid/antagonists & inhibitors , Hyaluronic Acid/metabolism , Acetylcholine/pharmacology , Animals , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Atherosclerosis/physiopathology , Blood Pressure/drug effects , Blood Pressure/physiology , Disease Models, Animal , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiopathology , Female , Glycocalyx/drug effects , Glycocalyx/metabolism , Hymecromone/analogs & derivatives , Hymecromone/pharmacology , Mice , Mice, Knockout , Vasodilation/drug effects , Vasodilation/physiology , Vasodilator Agents/pharmacology
5.
Cell Signal ; 22(3): 476-83, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19913094

ABSTRACT

Sphingosine-1-phosphate (S1P) regulates cell growth and survival, migration and adhesion in many cell types. S1P is generated by sphingosine kinases (SphKs), and dephosphorylated by phosphatases or cleaved by S1P lyase. Extracellular S1P activates specific G protein-coupled receptors while intracellular S1P can mobilize Ca2+ from thapsigargin-sensitive stores. Here, we have studied Ca2+ signalling in mouse embryonic fibroblasts (MEFs) deficient in S1P lyase. In these cells, S1P and sphingosine concentrations were elevated about 6-fold and 2-fold, respectively, as measured by liquid chromatography/tandem mass spectrometry. Measurements with fura-2-loaded cells in suspension revealed that resting [Ca2+]i was elevated and agonist-induced [Ca2+]i increases were augmented in S1P lyase-deficient MEFs both in the presence and absence of extracellular Ca2+. Importantly, [Ca2+]i increases and Ca2+ mobilization induced by the SERCA inhibitor, thapsigargin, were augmented, indicating enhanced Ca2+ storage in S1P lyase-deficient MEFs. Measurements with single cells expressing the calmodulin-based Ca2+ sensor, cameleon, revealed that at least two cell types could be distinguished in both MEF cell populations, one with a rapid and transient [Ca2+]i increase and the other with a slower and prolonged [Ca2+]i elevation upon stimulation with thapsigargin. The area under the time course of thapsigargin-induced [Ca2+]i increases, reflecting overall Ca2+ release, was significantly increased by more than 50% in both rapidly and slowly responding S1P lyase-deficient cells. It is concluded that elevated concentrations of S1P and/or sphingosine lead to enhanced Ca2+ storage and elevated basal [Ca2+]i. S1P metabolism thus plays a role not only in acute Ca2+ mobilization but also in long-term regulation of Ca2+ homeostasis.


Subject(s)
Aldehyde-Lyases/metabolism , Calcium/metabolism , Fibroblasts/metabolism , Aldehyde-Lyases/deficiency , Aldehyde-Lyases/genetics , Animals , Calcium Signaling , Calmodulin/metabolism , Cells, Cultured , Enzyme Inhibitors/pharmacology , Lysophospholipids/metabolism , Mice , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Thapsigargin/pharmacology
6.
Biochim Biophys Acta ; 1791(5): 357-70, 2009 May.
Article in English | MEDLINE | ID: mdl-19830907

ABSTRACT

Sphingosine-1-phosphate (S1P), formed by sphingosine kinases (SphKs), regulates cellular proliferation and migration by acting as an agonist at specific receptors or intracellularly. Since S1P's effects are probably dependent on subcellular localization of its formation and degradation, we have studied the influence of G protein-coupled receptors on the localization of SphK1. Activation of Gq-coupled receptors induced a profound, rapid (half-life 3-5 s) and long-lasting (> 2 h) translocation of SphK1 to the plasma membrane. This was mimicked by expression of constitutively active G protein alpha-subunits specifically of the Gq family. Classical Gq signalling pathways, or phosphorylation at Ser225, phospholipase D and Ca2+/calmodulin were not involved in M3 receptor-induced SphK1 translocation in HEK-293 cells. Translocation was associated with S1P receptor internalization, which was dependent on catalytic activity of SphK1 and S1P receptor binding and thus resulted from S1P receptor cross-activation. It is concluded that SphK1 is an important effector of Gq-coupled receptors, linking them via cross-activation of S1P receptors to G(i) and G12/13 signalling pathways.


Subject(s)
Cell Membrane/enzymology , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Receptors, Lysosphingolipid/metabolism , Animals , Cell Membrane/drug effects , Diglycerides/metabolism , Endocytosis/drug effects , Humans , Mice , Phospholipase D/metabolism , Phosphoserine/metabolism , Protein Kinase C/metabolism , Protein Transport/drug effects , Receptor, Muscarinic M3/metabolism , Sphingosine/analogs & derivatives , Sphingosine/pharmacology
7.
Basic Res Cardiol ; 104(1): 50-9, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18777003

ABSTRACT

Local formation of the sphingomyelin metabolite sphingosine-1-phosphate (S1P) within the vascular wall has been shown to modulate vascular reactivity. In this study we investigated whether sphingosine kinase, the enzyme responsible for S1P synthesis, plays a role in muscarinic receptor-mediated NO production and vascular relaxation in different blood vessel types. For this purpose, sphingosine kinase translocation and sphingolipid-dependent NO-production after muscarinic receptor stimulation were assessed in an endothelial cell line. Furthermore, we used the sphingosine kinase inhibitor N,N-dimethylsphingosine (DMS) to investigate the role of sphingosine kinase in the relaxant responses to the muscarinic agonist methacholine (MCh) in isolated rat aorta and mesenteric arteries. Activation of M(3)-receptors in an endothelial cell line induced a fast translocation of YFP-tagged sphingosine kinase-1 from the cytosol to the plasma membrane. Concomitant NO-production in this cell line was partially inhibited by DMS. Accordingly, in rat aorta the relaxant responses to MCh were attenuated in the presence of DMS, while the responses to the NO-donor sodium nitroprusside were unaltered. In contrast, DMS enhanced the relaxant responses to MCh in mesenteric artery preparations. This effect could also be observed in the presence of NO synthase and cyclooxygenase inhibitors, indicating that sphingosine kinase inhibition specifically enhanced endothelium-derived hyperpolarizing factor-mediated (i.e. non-NO and non-prostacyclin-dependent) relaxation. We conclude that sphingosine kinase differentially regulates vascular tone in different vessel types, enhancing NO-dependent vasorelaxation but counteracting EDHF-dependent vasorelaxation. This observation enhances our understanding of the complex mechanisms by which sphingolipids regulate vascular homeostasis. Moreover, a disturbed regulation of sphingolipid metabolism in the vascular wall may therefore play a role in the aetiology/pathology of disease states characterized by endothelial dysfunction.


Subject(s)
Biological Factors/physiology , Endothelium, Vascular/physiology , Enzyme Activation/physiology , Nitric Oxide/physiology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Receptors, Muscarinic/physiology , Vasodilation/physiology , Animals , Cerebrovascular Circulation/physiology , DNA Primers , Endothelium, Vascular/cytology , Genetic Markers , Luminescent Proteins/genetics , Mice , Phosphotransferases (Alcohol Group Acceptor)/genetics , Polymerase Chain Reaction , Protein Transport
8.
J Invest Dermatol ; 128(6): 1487-98, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18172456

ABSTRACT

The lysophospholipids, sphingosine-1-phosphate (S1P) and lysophosphatidic acid (LPA), stimulate chemotaxis and induce differentiation of human keratinocytes. As Ca(2+) plays an important role in keratinocyte differentiation, we studied Ca(2+) signaling by S1P and LPA in these cells, known to express mRNA transcripts of the S1P(1-5) and LPA(1-3) receptors, and the receptor subtypes involved in this process. S1P and LPA caused transient increases in intracellular free Ca(2+) concentration ([Ca(2+)](i)), with pEC(50) values of 8.5+/-0.11 and 7.5+/-0.23, respectively. The [Ca(2+)](i) increases are apparently mediated by stimulation of phospholipase C and involve Ca(2+) mobilization from thapsigargin-sensitive stores and subsequent Ca(2+) influx. The LPA-induced [Ca(2+)](i) increases were not inhibited by the LPA(1/3) receptor antagonist, dioctanoylglycerol pyrophosphate. The S1P-induced [Ca(2+)](i) increases were largely inhibited by the putative S1P(3) antagonist, BML-241, and the S1P(1/3) antagonist, VPC23019. The S1P(1)-specific agonist, SEW2871, did not increase [Ca(2+)](i) but stimulated chemotaxis of keratinocytes, which was fully blocked by S1P(1) antisense oligonucleotides. The data indicate that LPA and S1P potently increase [Ca(2+)](i) in human keratinocytes and that the effect of LPA is mediated by LPA(2), whereas that of S1P is mediated at least to a large part by S1P(3). The S1P(1) receptor, without stimulating [Ca(2+)](i) increases, mediates chemotaxis of keratinocytes.


Subject(s)
Calcium Signaling , Calcium/metabolism , Keratinocytes/cytology , Receptors, Lysophospholipid/metabolism , Cell Movement , Chemotaxis , Green Fluorescent Proteins/metabolism , Humans , Keratinocytes/metabolism , Lysophospholipids/metabolism , Models, Biological , Receptors, Lysosphingolipid/metabolism , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Thapsigargin/metabolism , Thiazolidines/pharmacology , Type C Phospholipases/metabolism
9.
J Eukaryot Microbiol ; 54(1): 42-4, 2007.
Article in English | MEDLINE | ID: mdl-17300518

ABSTRACT

Arginine biosynthesis in eukaryotes is divided between the mitochondria and the cytosol. The anaerobic chytridiomycete Neocallimastix frontalis contains highly reduced, anaerobic modifications of mitochondria, the hydrogenosomes. Hydrogenosomes also occur in the microaerophilic flagellate Trichomonas vaginalis, which does not produce arginine but uses one of the mitochondrial enzymes, ornithine transcarbamoylase, in a cytosolic arginine dihydrolase pathway for ATP generation. EST sequencing and analysis of the hydrogenosomal proteome of N. frontalis provided evidence for two mitochondrial enzymes of arginine biosynthesis, carbamoylphosphate synthase and ornithine transcarbamoylase, while activities of the arginine dehydrolase pathway enzymes were not detectable in this fungus.


Subject(s)
Arginine/biosynthesis , Neocallimastix/metabolism , Organelles/metabolism , Amino Acid Sequence , Carbamoyl-Phosphate Synthase (Ammonia)/analysis , Carbamoyl-Phosphate Synthase (Ammonia)/chemistry , DNA, Complementary , Expressed Sequence Tags , Fungal Proteins , Gene Library , Molecular Sequence Data , Neocallimastix/enzymology , Organelles/chemistry , Ornithine Carbamoyltransferase/analysis , Ornithine Carbamoyltransferase/chemistry , Proteome , Sequence Alignment
10.
Naunyn Schmiedebergs Arch Pharmacol ; 374(5-6): 413-28, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17242884

ABSTRACT

Sphingosine kinases (SphKs) catalyze the phosphorylation of sphingosine to sphingosine-1-phosphate (S1P). Together with other sphingolipid metabolizing enzymes, SphKs regulate the balance of the lipid mediators, ceramide, sphingosine, and S1P. The ubiquitous mediator S1P regulates cellular functions such as proliferation and survival, cytoskeleton architecture and Ca(2+) homoeostasis, migration, and adhesion by activating specific high-affinity G-protein-coupled receptors or by acting intracellularly. In mammals, two isoforms of SphK have been identified. They are activated by G-protein-coupled receptors, receptor tyrosine kinases, immunoglobulin receptors, cytokines, and other stimuli. The molecular mechanisms by which SphK1 and SphK2 are specifically regulated are complex and only partially understood. Although SphK1 and SphK2 appear to have opposing roles, promoting cell growth and apoptosis, respectively, they can obviously also substitute for each other, as mice deficient in either SphK1 or SphK2 had no obvious abnormalities, whereas double-knockout animals were embryonic lethal. In this review, our understanding of structure, regulation, and functional roles of SphKs is updated and discussed with regard to their implication in pathophysiological and disease states.


Subject(s)
Phosphotransferases (Alcohol Group Acceptor)/metabolism , Signal Transduction/physiology , Amino Acid Sequence , Animals , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Lysophospholipids/metabolism , Molecular Sequence Data , Phosphotransferases (Alcohol Group Acceptor)/genetics , Sequence Homology, Amino Acid , Sphingosine/analogs & derivatives , Sphingosine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...