Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Neuropharmacology ; 43(6): 966-78, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12423666

ABSTRACT

We report here that human Ntera-2/D1 (NT-2) cells, an undifferentiated committed neuronal progenitor cell line, endogenously express a functional P2Y(1) receptor, while other P2Y subtypes, except perhaps P2Y(4), are not functionally expressed. Quantitative RT-PCR analysis showed that NT-2 cells abundantly express mRNA for P2Y(1) and P2Y(11) receptors, while P2Y(2) and P2Y(4) receptors were detected at considerably lower levels. Western blot analysis also demonstrated expression of P2Y(1) receptors and Galpha(q/11) subunits. Various nucleotides induced intracellular Ca(2+) mobilisation in NT-2 cells in a concentration-dependent manner with a rank order potency of 2-MeSADP > 2-MeSATP > ADP > ATP > UTP > ATPgammaS, a profile resembling that of human P2Y(1) receptors. Furthermore, P2Y(1) receptor-specific (A3P5P) and P2Y-selective (PPADS, suramin) antagonists inhibited adenine nucleotide-induced Ca(2+) responses in a concentration-dependent manner, consistent with expression of a P2Y(1) receptor. Moreover, of seven adenine nucleotides tested, only Bz-ATP and ATPgammaS elicited small increases in cAMP formation suggesting that few, if any, functional P2Y(11) receptors were expressed. P2Y(1) receptor-selective adenine nucleotides, including 2-MeSADP and ADP, also induced concentration-dependent phosphorylation and hence, activation of the extracellular-signal regulated protein kinases (ERK1/2). NT-2 cells, therefore, provide a useful neuronal-like cellular model for studying the precise signalling pathways and physiological responses mediated by a native P2Y(1) receptor.


Subject(s)
Adenosine Diphosphate/analogs & derivatives , Adenosine Triphosphate/analogs & derivatives , Membrane Proteins , Neurons/metabolism , Pyridoxal Phosphate/analogs & derivatives , Receptors, Purinergic P2/metabolism , Stem Cells/metabolism , Adenosine Diphosphate/pharmacology , Adenosine Triphosphate/pharmacology , Antineoplastic Agents/pharmacology , Blotting, Western , Calcium/metabolism , Cell Membrane/metabolism , Cells, Cultured , Cyclic AMP , Fluorescence , GTP-Binding Protein alpha Subunits, Gq-G11 , Heterotrimeric GTP-Binding Proteins/metabolism , Humans , Mitogen-Activated Protein Kinases/drug effects , Mitogen-Activated Protein Kinases/metabolism , Platelet Aggregation Inhibitors/pharmacology , Pyridoxal Phosphate/pharmacology , RNA, Messenger/analysis , Receptors, Purinergic P2/classification , Receptors, Purinergic P2Y1 , Receptors, Purinergic P2Y12 , Receptors, Purinergic P2Y2 , Reverse Transcriptase Polymerase Chain Reaction/methods , Suramin/pharmacology , Thionucleotides/pharmacology , Uridine Triphosphate/pharmacology
2.
Biochim Biophys Acta ; 1521(1-3): 107-19, 2001 Oct 31.
Article in English | MEDLINE | ID: mdl-11690642

ABSTRACT

The diverse biological actions of extracellular nucleotides in tissues and cells are mediated by two distinct classes of P2 receptor, P2X and P2Y. The G protein-coupled P2Y receptors comprise at least six mammalian subtypes (P2Y(1,2,4,6,11,12)), all of which have been cloned from human tissues, as well as other species. The P2Y receptor subtypes differ in their pharmacological selectivity for various adenosine and uridine nucleotides, which overlap in some cases. Data concerning the mRNA expression patterns of five P2Y receptors (P2Y(1,2,4,6,11)) in different human tissues and cells are currently quite limited, while P2Y mRNA distribution in the human brain has not previously been studied. In this study, we have addressed this deficiency in receptor expression data by using a quantitative reverse transcription-polymerase chain reaction approach to measure the precise mRNA expression pattern of each P2Y receptor subtype in a number of human peripheral tissues and brain regions, from multiple individuals, as well as numerous human cell lines and primary cells. All five P2Y receptors exhibited widespread yet subtype-selective mRNA expression profiles throughout the human tissues, brain regions and cells used. Our extensive expression data indicate the many potentially important roles of P2Y receptors throughout the human body, and will help in elucidating the physiological function of each receptor subtype in a wide variety of human systems.


Subject(s)
Polymerase Chain Reaction/methods , RNA, Messenger/metabolism , Receptors, Purinergic P2/metabolism , Actins/analysis , Brain/metabolism , Cell Line , Cyclophilins/analysis , DNA Probes , Female , Glyceraldehyde-3-Phosphate Dehydrogenases/analysis , Humans , Male , Protein Isoforms/analysis , Protein Isoforms/metabolism , RNA, Messenger/analysis , Receptors, Purinergic P2/analysis , Reverse Transcriptase Polymerase Chain Reaction
3.
Biochem J ; 359(Pt 3): 507-16, 2001 Nov 01.
Article in English | MEDLINE | ID: mdl-11672424

ABSTRACT

GSH-dependent prostaglandin D(2) synthase (PGDS) enzymes represent the only vertebrate members of class Sigma glutathione S-transferases (GSTs) identified to date. Complementary DNA clones encoding the orthologous human and rat GSH-dependent PGDS (hPGDS and rPGDS, respectively) have been expressed in Escherichia coli, and the recombinant proteins isolated by affinity chromatography. The purified enzymes were both shown to catalyse specifically the isomerization of prostaglandin (PG) H(2) to PGD(2). Each transferase also exhibited GSH-conjugating and GSH-peroxidase activities. The ability of hPGDS to catalyse the conjugation of aryl halides and isothiocyanates with GSH was found to be less than that of the rat enzyme. Whilst there is no difference between the enzymes with respect to their K(m) values for 1-chloro-2,4-dinitrobenzene, marked differences were found to exist with respect to their K(m) for GSH (8 mM versus 0.3 mM for hPGDS and rPGDS, respectively). Using molecular modelling techniques, amino acid substitutions have been identified in the N-terminal domain of these enzymes that lie outside the proposed GSH-binding site, which may explain these catalytic differences. The tissue-specific expression of PGDS also varies significantly between human and rat; amongst the tissues examined, variation in expression between the two species was most apparent in spleen and bone marrow. Differences in catalytic properties and tissue-specific expression of hPGDS and rPGDS appears to reflect distinct physiological roles for class Sigma GST between species. The evolution of divergent functions for the hPGDS and rPGDS is discussed in the context of the orthologous enzyme from chicken.


Subject(s)
Glutathione Transferase/metabolism , Intramolecular Oxidoreductases/metabolism , Amino Acid Sequence , Animals , Catalysis , Glutathione Transferase/classification , Glutathione Transferase/genetics , Humans , Intramolecular Oxidoreductases/chemistry , Intramolecular Oxidoreductases/genetics , Isoenzymes/metabolism , Lipocalins , Models, Molecular , Molecular Sequence Data , Organ Specificity , Protein Conformation , Protein Structure, Tertiary , Rats , Rats, Wistar , Recombinant Proteins/metabolism , Sequence Alignment
4.
Immunology ; 103(3): 319-31, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11454061

ABSTRACT

The presentation of extremely low doses of antigen to T cells is enhanced by immunoglobulin E (IgE)-dependent antigen focusing to CD23, the low-affinity receptor for IgE, expressed on activated B cells. CD23 contains a C-type lectin domain in its extracellular sequence and a targeting signal for coated pits, required for endocytosis, in its cytoplasmic sequence. CD23 is non-covalently associated with the major histocompatibility complex class II antigen, human leucocyte antigen HLA-DR, on the surface of human B cells, but the fate of this complex following endocytosis is unknown. To answer this question we have labelled these proteins on the surface of RPMI 8866 B cells and traced their route through the cytoplasm. Endocytosis mediated by anti-CD23 antibodies (BU38 and MHM6) led to the loss of CD23 from the cells. Endocytosis mediated by an antibody to HLA-DR (CR3/43) or an antigen-IgE complex (NP-BSA-anti-NP IgE), however, led to recycling of the HLA-DR-CD23 complex to the cell surface on a time scale (3-6 hr) consistent with the recycling of HLA-DR in antigen presentation. Along the latter pathway CD23 label was observed in cytoplasmic organelles that resembled the 'compartments for peptide loading' or 'class II vesicles' described by previous authors. Two features of the recycling process may contribute to the efficiency of antigen presentation. Peptide exchange may be facilitated by the proximity of HLA-DR and antigen in peptide loading compartments of the endosomal network. The return of CD23 with HLA-DR to the cell surface may then help to stabilize specific B-cell-T-cell interactions, contributing to T-cell activation.


Subject(s)
B-Lymphocytes/immunology , Endocytosis/immunology , HLA-DR Antigens/metabolism , Receptors, IgE/metabolism , Antibodies, Monoclonal/immunology , Antigen-Antibody Complex/immunology , B-Lymphocytes/ultrastructure , Cell Culture Techniques , Cytoplasmic Vesicles/immunology , Electrophoresis, Polyacrylamide Gel , Endosomes/immunology , Endosomes/ultrastructure , HLA-DR Antigens/immunology , Humans , Immunoglobulin E/immunology , Microscopy, Confocal , Microscopy, Electron , Receptors, IgE/immunology
5.
Brain Res ; 892(1): 94-101, 2001 Feb 16.
Article in English | MEDLINE | ID: mdl-11172753

ABSTRACT

We have cloned and functionally expressed the human orthologue of the mouse TRAAK gene. When cDNA for hTRAAK is expressed in either Xenopus oocytes or HEK293 cells it forms a K(+)-selective conductance and hyperpolarises the resting membrane potential. Quantitative mRNA expression analysis using Taqman revealed that hTRAAK mRNA is predominantly present in the central nervous system where it exhibits a regionally diverse pattern of expression. Like the related channel TREK-1, the activity of TRAAK was potentiated by arachidonic acid. The neuroprotective agent sipatrigine (10 microM) inhibited both hTREK-1 (73.3+/-4.4%) and hTRAAK (45.1+/-11.2%) in a reversible, voltage-independent manner. Inhibition of both channels was dose-dependent and for TREK-1 occurred with an IC(50) of 4 microM. The related compound lamotrigine, which is a better anticonvulsant but weaker neuroprotective agent than sipatrigine, was a far less effective antagonist of both channels, producing <10% inhibition at a concentration of 10 microM.


Subject(s)
Brain/physiology , Neuroprotective Agents/pharmacology , Piperazines/pharmacology , Potassium Channels, Tandem Pore Domain , Potassium Channels/physiology , Pyrimidines/pharmacology , Amino Acid Sequence , Animals , Cell Line , Female , Humans , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Molecular Sequence Data , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/physiology , Oocytes/drug effects , Oocytes/physiology , Potassium Channel Blockers , Potassium Channels/chemistry , Potassium Channels/genetics , RNA, Messenger/genetics , Sequence Alignment , Sequence Homology, Amino Acid , Transfection , Xenopus laevis
6.
Osteoarthritis Cartilage ; 9(8): 727-37, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11795992

ABSTRACT

OBJECTIVE: To characterize the expression pattern of clusterin in adult human normal and osteoarthritic cartilage. METHODS: Clusterin mRNA expression in adult human normal and osteoarthritic cartilage was investigated by analysis of cDNA libraries, TaqMan quantitative RT-PCR, microarray and in situ hybridization. RESULTS: Sequence analysis of ESTs from adult human normal and osteoarthritic cartilage cDNA libraries demonstrated that the abundance of clusterin in these libraries was equivalent to genes which have been more commonly associated with cartilage. To examine tissue distribution, TaqMan Quantitative PCR analysis was performed using RNA from a panel of individual normal tissues. Clusterin was expressed at significant levels in cartilage, brain, liver, and pancreas. The expression of clusterin mRNA was up-regulated in early osteoarthritic vs normal cartilage when analysed by microarray analysis. Using in situ hybridization, chondrocytes of normal cartilage expressed moderate levels of clusterin. Upper mid-zone chondrocytes in cartilage with early stages of osteoarthritic disease expressed high levels of clusterin mRNA. In advanced osteoarthritic cartilage, the overall expression of clusterin was reduced. CONCLUSION: The induction of clusterin has been associated with a variety of disease states where it appears to provide a cytoprotective effect. The increased expression of clusterin mRNA in the early stages of osteoarthritis (OA) may reflect an attempt by the chondrocytes to protect and repair the tissue. In contrast, the decrease in clusterin mRNA in the advanced osteoarthritic cartilage accompanies the final degenerative stages of the disease. An understanding of the expression of clusterin in osteoarthritis may allow consideration of this protein as a marker for cartilage changes in this chronic degenerative condition.


Subject(s)
Cartilage, Articular/metabolism , Glycoproteins/metabolism , Molecular Chaperones/metabolism , Osteoarthritis/metabolism , Adult , Aged , Aged, 80 and over , Case-Control Studies , Clusterin , Female , Gene Library , Humans , In Situ Hybridization/methods , Male , Middle Aged , Oligonucleotide Array Sequence Analysis/methods , Proteoglycans/metabolism , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation
7.
Mol Cell Neurosci ; 16(5): 609-19, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11083922

ABSTRACT

Sequential proteolytic processing of the Amyloid Precursor Protein (APP) by beta- and gamma-secretases generates the 4-kDa amyloid (A beta) peptide, a key component of the amyloid plaques seen in Alzheimer's disease (AD). We and others have recently reported the identification and characterisation of an aspartic proteinase, Asp2 (BACE), as beta-secretase. Here we describe the characterization of a second highly related aspartic proteinase, Asp1 as a second beta-secretase candidate. Asp1 is expressed in brain as detected at the mRNA level and at the protein level. Transient expression of Asp1 in APP-expressing cells results in an increase in the level of beta-secretase-derived soluble APP and the corresponding carboxy-terminal fragment. Paradoxically there is a decrease in the level of soluble A beta secreted from the cells. Asp1 colocalizes with APP in the Golgi/endoplasmic reticulum compartments of cultured cells. Asp1, when expressed as an Fc fusion protein (Asp1-Fc), has the N-terminal sequence ALEP..., indicating that it has lost the prodomain. Asp1-Fc exhibits beta-secretase activity by cleaving both wild-type and Swedish variant (KM/NL) APP peptides at the beta-secretase site.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Aspartic Acid Endopeptidases/metabolism , Glycoproteins/genetics , Glycoproteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Amyloid Precursor Protein Secretases , Amyloid beta-Protein Precursor/analysis , Amyloid beta-Protein Precursor/chemistry , Animals , Aspartic Acid Endopeptidases/chemistry , Binding Sites/physiology , COS Cells , Cloning, Molecular , Endopeptidases , Female , Glycoproteins/analysis , Humans , Male , Membrane Proteins/analysis , Molecular Sequence Data , Rabbits , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid
8.
Brain Res Mol Brain Res ; 82(1-2): 74-83, 2000 Oct 20.
Article in English | MEDLINE | ID: mdl-11042359

ABSTRACT

We have isolated, by degenerate PCR, a complementary DNA encoding a novel two pore domain potassium channel. This is the 7th functional member of the human tandem pore domain potassium channel family to be reported. It has an open reading frame of 1.125 kb and encodes a 374 amino acid protein which shows 62% identity to the human TASK-1 gene: identity to other human members of the family is 31-35% at the amino acid level. We believe this gene to be human TASK-3, the ortholog of the recently reported rat TASK-3 gene: amino acid identity between the two is 74%. 'Taqman' mRNA analysis demonstrated a very specific tissue distribution pattern, showing human TASK-3 mRNA to be localised largely in the cerebellum, in contrast rat TASK-3 was reported to be widely distributed. We have shown by radiation hybrid mapping that human TASK-3 can be assigned to chromosome 8q24.3. Human TASK-3 was demonstrated to endow Xenopus oocytes with a negative resting membrane potential through the presence of a large K(+) selective conductance. TASK-3 is inhibited by extracellular acidosis with a mid-point of inhibition around pH 6. 5, supporting the predictions from the sequence data that this is a third human TASK (TWIK-related acid sensitive K(+) channel) gene.


Subject(s)
Cerebellum/metabolism , Chromosomes, Human, Pair 8 , Evoked Potentials/physiology , Nerve Tissue Proteins , Potassium Channels, Tandem Pore Domain , Potassium Channels/genetics , Amino Acid Sequence , Animals , Base Sequence , Chromosome Mapping , Cloning, Molecular , DNA, Complementary , Genetic Variation , Humans , Membrane Potentials/physiology , Molecular Sequence Data , Oocytes/physiology , Phylogeny , Polymerase Chain Reaction , Potassium Channels/chemistry , Potassium Channels/physiology , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid
9.
Biochem Biophys Res Commun ; 273(3): 805-10, 2000 Jul 14.
Article in English | MEDLINE | ID: mdl-10891327

ABSTRACT

We have identified a cDNA, designated HOFNH30, which encodes a 354 amino acid G-protein-coupled receptor (GPCR). This receptor has 96% amino acid identity to the Jurkat-T cell-derived EDG7 and could be a splice variant. RT-PCR analysis demonstrated that HOFNH30 mRNA is expressed in placenta whereas EDG7 mRNA shows highest expression in prostate. The HOFNH30 gene is localized to human chromosome 1p22. 3-1p31.1. When HOFNH30 was expressed in RBL-2H3 cells, LPA and phosphatidic acid (PA) induced a calcium mobilization response with EC(50) values of 13 nM and 3 microM, respectively. LPA also induced phosphorylation of mitogen-activated protein kinase (p42(MAPK) and p44(MAPK)) in HOFNH30-transfected but not vector-transfected RBL-2H3 cells. In the present study, we have identified a novel variant from the EDG receptor family, a GPCR for which LPA is a high-affinity endogenous ligand.


Subject(s)
GTP-Binding Proteins/metabolism , Lysophospholipids/metabolism , Receptors, Cell Surface/genetics , Receptors, G-Protein-Coupled , Amino Acid Sequence , Base Sequence , Calcium/metabolism , Chromosome Mapping , Chromosomes, Human, Pair 1 , Cloning, Molecular , Enzyme Activation , Humans , Jurkat Cells , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Molecular Sequence Data , Phosphorylation , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/metabolism , Receptors, Lysophosphatidic Acid , Sequence Homology, Amino Acid , Tumor Cells, Cultured
10.
J Allergy Clin Immunol ; 105(6 Pt 1): 1093-100, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10856141

ABSTRACT

BACKGROUND: Eosinophils, basophils, and mast cells are believed to be the central tenet cells in allergic conditions including allergic rhinitis, asthma, and eczema. The molecular mechanisms underlying the recruitment of these cells to sites of allergic inflammation are poorly understood. OBJECTIVES: Our aim was to identify a common adhesion molecule that could potentially be responsible for mediating the recruitment of the allergic cell types to the lungs and other sites of allergy. METHODS: We have cloned a sialoadhesin molecule from a human eosinophil library with the use of expressed sequence tag technology and characterized its expression on allergic cells by the use of flow cytometry and specific mAbs. RESULTS: With the use of expressed sequence tag sequencing, we have identified a novel siglec molecule, SAF-2. SAF-2 has homology with other sialoadhesin family members (CD33 and siglec-5) and belongs to a subgroup of the Ig superfamily. SAF-2 is a 431-amino acid protein composed of 3 Ig domains with a 358-amino acid extracellular domain and a 47-amino acid tail. SAF-2 is highly restricted to eosinophils, basophils, and mast cells. Antibodies to SAF-2 do not modulate Ca(++) mobilization or chemotaxis of human eosinophils induced by eotaxin. CONCLUSION: SAF-2 is a highly restricted sialoadhesin molecule, which may be useful in the detection and/or modulation of allergic cells.


Subject(s)
Antigens, CD/biosynthesis , Antigens, Differentiation, B-Lymphocyte/biosynthesis , Antigens, Surface/biosynthesis , Basophils/metabolism , Eosinophils/metabolism , Hypersensitivity/pathology , Lectins , Mast Cells/metabolism , Antigens, CD/genetics , Antigens, CD/physiology , Antigens, Differentiation, B-Lymphocyte/genetics , Antigens, Differentiation, B-Lymphocyte/physiology , Antigens, Surface/genetics , Antigens, Surface/physiology , Erythrocytes/metabolism , Gene Expression , Humans , N-Acetylneuraminic Acid/pharmacology , RNA, Messenger/genetics , Sequence Homology, Amino Acid
11.
J Biol Chem ; 275(34): 25965-71, 2000 Aug 25.
Article in English | MEDLINE | ID: mdl-10851242

ABSTRACT

Opiate tolerance and dependence are major clinical and social problems. The anti-opiate neuropeptides FF and AF (NPFF and NPAF) have been implicated in pain modulation as well as in opioid tolerance and may play a critical role in this process, although their mechanism of action has remained unknown. Here we describe a cDNA encoding a novel neuropeptide Y-like human orphan G protein-coupled receptor (GPCR), referred to as HLWAR77 for which NPAF and NPFF have high affinity. Cells transiently or stably expressing HLWAR77 bind and respond in a concentration-dependent manner to NPAF and NPFF and are also weakly activated by FMRF-amide (Phe-Met-Arg-Phe-amide) and a variety of related peptides. The high affinity and potency of human NPFF and human NPAF for HLWAR77 strongly suggest that these are the cognate ligands for this receptor. Expression of HLWAR77 was demonstrated in brain regions associated with opiate activity, consistent with the pain-modulating activity of these peptides, whereas the expression in adipose tissue suggests other physiological and pathophysiological activities for FMRF-amide neuropeptides. The discovery that the anti-opiate neuropeptides are the endogenous ligands for HLWAR77 will aid in defining the physiological role(s) of these ligands and facilitate the identification of receptor agonists and antagonists.


Subject(s)
Neuropeptides/metabolism , Oligopeptides/metabolism , Receptors, Neuropeptide/metabolism , Amino Acid Sequence , Arrestins/metabolism , Base Sequence , Calcium/metabolism , Cell Line , FMRFamide/pharmacology , Humans , Ligands , Molecular Sequence Data , Receptors, Neuropeptide/genetics , beta-Arrestins
12.
J Biol Chem ; 275(27): 20247-50, 2000 Jul 07.
Article in English | MEDLINE | ID: mdl-10811630

ABSTRACT

Neuromedins are a family of peptides best known for their contractile activity on smooth muscle preparations. The biological mechanism of action of neuromedin U remains unknown, despite the fact that the peptide was first isolated in 1985. Here we show that neuromedin U potently activates the orphan G protein-coupled receptor FM3, with subnanomolar potency, when FM3 is transiently expressed in human HEK-293 cells. Neuromedins B, C, K, and N are all inactive at this receptor. Quantitative reverse transcriptase-polymerase chain reaction analysis of neuromedin U expression in a range of human tissues showed that the peptide is highly expressed in the intestine, pituitary, and bone marrow, with lower levels of expression seen in stomach, adipose tissue, lymphocytes, spleen, and the cortex. Similar analysis of FM3 expression showed that the receptor is widely expressed in human tissue with highest levels seen in adipose tissue, intestine, spleen, and lymphocytes, suggesting that neuromedin U may have a wide range of presently undetermined physiological effects. The discovery that neuromedin U is an endogenous agonist for FM3 will significantly aid the study of the full physiological role of this peptide.


Subject(s)
GTP-Binding Proteins/metabolism , Membrane Proteins , Neuropeptides/pharmacology , Receptors, Cell Surface/agonists , Receptors, Neurotransmitter , Calcium/metabolism , Cell Line , Cloning, Molecular , Gene Expression Regulation , Humans , Inositol Phosphates/metabolism , Neuropeptides/genetics , RNA, Messenger/analysis , RNA, Messenger/metabolism , Receptors, Cell Surface/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transfection
13.
Br J Pharmacol ; 128(1): 1-3, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10498827

ABSTRACT

The cellular mechanisms underlying the physiological effects of the orexins are poorly understood. Therefore, the pharmacology of the recombinant human orexin receptors was studied using FLIPR. Intracellular calcium ([Ca2+]i) was monitored in Chinese hamster ovary (CHO) cells stably expressing orexin-1 (OX1) or orexin-2 (OX2) receptors using Fluo-3AM. Orexin-A and orexin-B increased [Ca2+]i in a concentration dependent manner in CHO-OX1 (pEC50=8.03+/-0.08 and 7. 30+/-0.08 respectively, n=5) and CHO-OX2 (pEC50=8.18+/-0.10 and 8. 43+/-0.09 respectively, n=5) cells. This response was typified as a rapid peak in [Ca2+]i (maximal at 6 - 8 s), followed by a gradually declining secondary phase. Thapsigargin (3 microM) or U73122 (3 microM) abolished the response. In calcium-free conditions the peak response was unaffected but the secondary phase was shortened, returning to basal values within 90 s. Calcium (1.5 mM) replacement restored the secondary phase. In conclusion, orexins cause a phospholipase C-mediated release of calcium from intracellular stores, with subsequent calcium influx.


Subject(s)
Calcium/metabolism , Carrier Proteins/pharmacology , Intracellular Signaling Peptides and Proteins , Neuropeptides/pharmacology , Receptors, Neuropeptide/metabolism , Aniline Compounds , Animals , CHO Cells , Calcium/antagonists & inhibitors , Calcium/pharmacology , Calcium Signaling/drug effects , Carrier Proteins/antagonists & inhibitors , Cricetinae , Dose-Response Relationship, Drug , Fluorescent Dyes , Humans , Neuropeptides/antagonists & inhibitors , Orexin Receptors , Orexins , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phospholipase D/antagonists & inhibitors , Phospholipase D/metabolism , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Receptors, G-Protein-Coupled , Receptors, Neuropeptide/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Thapsigargin/pharmacology , Time Factors , Type C Phospholipases/antagonists & inhibitors , Type C Phospholipases/metabolism , Xanthenes
14.
Mol Pharmacol ; 56(3): 657-63, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10462554

ABSTRACT

The cysteinyl leukotrienes (CysLTs) have been implicated in the pathophysiology of inflammatory disorders, in particular asthma, for which the CysLT receptor antagonists pranlukast, zafirlukast, and montelukast, have been introduced recently as novel therapeutics. Here we report on the molecular cloning, expression, localization, and pharmacological characterization of a CysLT receptor (CysLTR), which was identified by ligand fishing of orphan seven-transmembrane-spanning, G protein-coupled receptors. This receptor, expressed in human embryonic kidney (HEK)-293 cells responded selectively to the individual CysLTs, LTC(4), LTD(4), or LTE(4), with a calcium mobilization response; the rank order potency was LTD(4) (EC(50) = 2.5 nM) > LTC(4) (EC(50) = 24 nM) > LTE(4) (EC(50) = 240 nM). Evidence was provided that LTE(4) is a partial agonist at this receptor. [(3)H]LTD(4) binding and LTD(4)-induced calcium mobilization in HEK-293 cells expressing the CysLT receptor were potently inhibited by the structurally distinct CysLTR antagonists pranlukast, montelukast, zafirlukast, and pobilukast; the rank order potency was pranlukast = zafirlukast > montelukast > pobilukast. LTD(4)-induced calcium mobilization in HEK-293 cells expressing the CysLT receptor was not affected by pertussis toxin, and the signal appears to be the result of the release from intracellular stores. Localization studies indicate the expression of this receptor in several tissues, including human lung, human bronchus, and human peripheral blood leukocytes. The discovery of this receptor, which has characteristics of the purported CysLT(1) receptor subtype, should assist in the elucidation of the pathophysiological roles of the CysLTs and in the identification of additional receptor subtypes.


Subject(s)
Membrane Proteins , Receptors, Leukotriene/genetics , Amino Acid Sequence , Base Sequence , Binding Sites , Biological Transport/drug effects , Calcium/metabolism , Cells, Cultured , Cloning, Molecular , Humans , Leukotriene D4/pharmacology , Molecular Sequence Data , Pertussis Toxin , Receptors, Leukotriene/metabolism , Signal Transduction/drug effects , Virulence Factors, Bordetella/pharmacology
17.
Br J Pharmacol ; 125(7): 1387-92, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9884064

ABSTRACT

The pharmaceutical industry has readily embraced genomics to provide it with new targets for drug discovery. Large scale DNA sequencing has allowed the identification of a plethora of DNA sequences distantly related to known G protein-coupled receptors (GPCRs), a superfamily of receptors that have a proven history of being excellent therapeutic targets. In most cases the extent of sequence homology is insufficient to assign these 'orphan' receptors to a particular receptor subfamily. Consequently, reverse molecular pharmacological and functional genomic strategies are being employed to identify the activating ligands of the cloned receptors. Briefly, the reverse molecular pharmacological methodology includes cloning and expression of orphan GPCRs in mammalian cells and screening these cells for a functional response to cognate or surrogate agonists present in biological extract preparations, peptide libraries, and complex compound collections. The functional genomics approach involves the use of 'humanized yeast cells, where the yeast GPCR transduction system is engineered to permit functional expression and coupling of human GPCRs to the endogenous signalling machinery. Both systems provide an excellent platform for identifying novel receptor ligands. Once activating ligands are identified they can be used as pharmacological tools to explore receptor function and relationship to disease.


Subject(s)
Drug Design , GTP-Binding Proteins/metabolism , Receptors, Cell Surface/metabolism , Animals , Drug Evaluation, Preclinical , Drug Industry , Humans , Peptides/pharmacology , Receptors, Calcitonin Gene-Related Peptide/metabolism , Receptors, Cell Surface/drug effects
18.
Eur J Immunol ; 27(12): 3228-35, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9464810

ABSTRACT

CD23, the low-affinity IgE receptor, is up-regulated on interleukin (IL)-4-stimulated B cells and monocytes, with a concomitant increase in the release of soluble fragments of CD23 (sCD23) into the medium by proteolytic processing of the surface-bound intact CD23. The effect of inhibition of the processing of CD23 on IgE production in human and mouse cells and in a mouse model in vivo was evaluated. CD23 processing to sCD23 from RPMI 8866 (a human Epstein-Barr virus-transformed B cell line) cell membranes was inhibited by a broad-spectrum matrix-metalloprotease inhibitor, batimastat, with an IC50 of 0.15 microM. Batimastat also inhibited CD23 processing in whole RPMI 8866 cells as well as in IL-4-stimulated purified human monocytes with similar IC50. Batimastat inhibited IgE production from IL-4/anti-CD40-stimulated human tonsil B cells as well as mouse splenic B cells in a manner consistent with inhibition of CD23 processing. Release of soluble fragments of CD23 in the cell supernatants of tonsil B cells was inhibited over the concentration range of 1-10 microM batimastat and intact cell surface CD23 was increased on mouse splenic B cells in the presence of these concentrations of batimastat. IgE production of IL-4-stimulated human peripheral blood mononuclear cells was also blocked by 1-10 microM batimastat, again with comparable inhibition of sCD23 release over the same concentration range. Finally, in a mouse model of IgE production, batimastat inhibited IgE production in response to ovalbumin challenge as determined by serum IgE levels. Taken together, the data support a role of CD23 in IgE production and point to CD23 processing to sCD23 as a therapeutically relevant control point in the regulation of IgE synthesis.


Subject(s)
B-Lymphocytes/immunology , Monocytes/immunology , Phenylalanine/analogs & derivatives , Protease Inhibitors/pharmacology , Receptors, IgE/immunology , Signal Transduction/immunology , Thiophenes/pharmacology , Animals , Cell Line, Transformed , Humans , Mice , Phenylalanine/pharmacology , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...