Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
Add more filters










Publication year range
1.
Nano Lett ; 16(12): 7842-7848, 2016 12 14.
Article in English | MEDLINE | ID: mdl-27960518

ABSTRACT

We report on a nanoinfrared (IR) imaging study of ultraconfined plasmonic hotspots inside graphene nanobubbles formed in graphene/hexagonal boron nitride (hBN) heterostructures. The volume of these plasmonic hotspots is more than one-million-times smaller than what could be achieved by free-space IR photons, and their real-space distributions are controlled by the sizes and shapes of the nanobubbles. Theoretical analysis indicates that the observed plasmonic hotspots are formed due to a significant increase of the local plasmon wavelength in the nanobubble regions. Such an increase is attributed to the high sensitivity of graphene plasmons to its dielectric environment. Our work presents a novel scheme for plasmonic hotspot formation and sheds light on future applications of graphene nanobubbles for plasmon-enhanced IR spectroscopy.

2.
J Immunol ; 166(10): 6341-8, 2001 May 15.
Article in English | MEDLINE | ID: mdl-11342658

ABSTRACT

The anaphylatoxin C3a is a potent chemotactic peptide and inflammatory mediator released during complement activation which binds to and activates a G-protein-coupled receptor. Molecular cloning of the C3aR has facilitated studies to identify nonpeptide antagonists of the C3aR. A chemical lead that selectively inhibited the C3aR in a high throughput screen was identified and chemically optimized. The resulting antagonist, N(2)-[(2,2-diphenylethoxy)acetyl]-L-arginine (SB 290157), functioned as a competitive antagonist of (125)I-C3a radioligand binding to rat basophilic leukemia (RBL)-2H3 cells expressing the human C3aR (RBL-C3aR), with an IC(50) of 200 nM. SB 290157 was a functional antagonist, blocking C3a-induced C3aR internalization in a concentration-dependent manner and C3a-induced Ca(2+) mobilization in RBL-C3aR cells and human neutrophils with IC(50)s of 27.7 and 28 nM, respectively. SB 290157 was selective for the C3aR in that it did not antagonize the C5aR or six other chemotactic G protein-coupled receptors. Functional antagonism was not solely limited to the human C3aR; SB 290157 also inhibited C3a-induced Ca(2+) mobilization of RBL-2H3 cells expressing the mouse and guinea pig C3aRS: It potently inhibited C3a-mediated ATP release from guinea pig platelets and inhibited C3a-induced potentiation of the contractile response to field stimulation of perfused rat caudal artery. Furthermore, in animal models, SB 290157, inhibited neutrophil recruitment in a guinea pig LPS-induced airway neutrophilia model and decreased paw edema in a rat adjuvant-induced arthritis model. This selective antagonist may be useful to define the physiological and pathophysiological roles of the C3aR.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Arginine/pharmacology , Benzhydryl Compounds/pharmacology , Complement C3a/metabolism , Complement Inactivator Proteins/pharmacology , Membrane Proteins , Receptors, Complement/antagonists & inhibitors , Animals , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Arginine/analogs & derivatives , Arginine/metabolism , Arginine/pharmacokinetics , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Benzhydryl Compounds/metabolism , Benzhydryl Compounds/pharmacokinetics , Binding, Competitive , Cell Line , Complement Inactivator Proteins/metabolism , Complement Inactivator Proteins/pharmacokinetics , Disease Models, Animal , Edema/pathology , Edema/prevention & control , Guinea Pigs , Hindlimb , Humans , Injections, Intraperitoneal , Leukocytosis/immunology , Leukocytosis/pathology , Male , Mice , Muscle Contraction/drug effects , Neutrophil Infiltration/drug effects , Rats , Rats, Inbred Lew , Rats, Sprague-Dawley , Receptors, Complement/metabolism , Tumor Cells, Cultured
3.
Eur J Pharmacol ; 413(2-3): 143-50, 2001 Feb 16.
Article in English | MEDLINE | ID: mdl-11226387

ABSTRACT

Starting with a partial sequence from Genbank, polymerase chain reaction (PCR) was utilized to isolate the full-length cDNA for NK(3) receptor from mouse brain. The murine NK(3) receptor has a predicted sequence of 452 amino acids, sharing 96% and 86% identity to the rat and human NK(3) receptors, respectively. Binding affinities and functional potencies of tachykinin receptor agonists were similar in HEK (human embryonic kidney) 293 cells expressing murine NK(3) receptor and human NK(3) receptor, although substance P and neurokinin A were more potent stimulators of Ca(2+) mobilization in murine NK(3) receptor cells. NK(3) receptor-selective antagonists from two structural classes, had 10- to 100-fold lower binding affinities for murine NK(3) receptor compared to human NK(3) receptor, and about 5- to 10-fold reduced potency in the murine NK(3) receptor functional assay. The results demonstrate species differences in the potencies of tachykinin receptor antagonists in murine and human NK(3) receptors, and the lower potencies in the former should be taken into consideration when using murine disease models.


Subject(s)
Calcium/metabolism , Receptors, Neurokinin-3/physiology , Amino Acid Sequence/physiology , Animals , Cloning, Molecular/methods , Humans , Mice , Molecular Sequence Data , Neurokinin A/metabolism , Neurokinin A/pharmacology , Quinolines/chemistry , Quinolines/metabolism , Quinolines/pharmacology , Receptors, Neurokinin-3/drug effects
4.
J Pharmacol Exp Ther ; 295(1): 373-81, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10992004

ABSTRACT

The pharmacological and pharmacokinetic profile of SB-222200 [(S)-(-)-N-(alpha-ethylbenzyl)-3-methyl-2-phenylquinoline-4-car boxami de], a human NK-3 receptor (hNK-3R) antagonist, was determined. SB-222200 inhibited (125)I-[MePhe(7)]neurokinin B (NKB) binding to Chinese hamster ovary (CHO) cell membranes stably expressing the hNK-3 receptor (CHO-hNK-3R) with a K(i) = 4.4 nM and antagonized NKB-induced Ca(2+) mobilization in HEK 293 cells stably expressing the hNK-3 receptor (HEK 293-hNK-3R) with an IC(50) = 18.4 nM. SB-222200 was selective for hNK-3 receptors compared with hNK-1 (K(i) > 100,000 nM) and hNK-2 receptors (K(i) = 250 nM). In HEK 293 cells transiently expressing murine NK-3 receptors (HEK 293-mNK-3R), SB-222200 inhibited binding of (125)I-[MePhe(7)]NKB (K(i) = 174 nM) and antagonized NKB (1 nM)-induced calcium mobilization (IC(50) = 265 nM). In mice oral administration of SB-222200 produced dose-dependent inhibition of behavioral responses induced by i.p. or intracerebral ventricular administration of the NK-3 receptor-selective agonist, senktide, with ED(50) values of approximately 5 mg/kg. SB-222200 effectively crossed the blood-brain barrier in the mouse and rat. The inhibitory effect of SB-222200 against senktide-induced behavioral responses in the mouse correlated significantly with brain, but not plasma, concentrations of the compound. Pharmacokinetic evaluation of SB-222200 in rat after oral administration (8 mg/kg) indicated sustained plasma concentrations (C(max) = about 400 ng/ml) and bioavailability of 46%. The preclinical profile of SB-222200, demonstrating high affinity, selectivity, reversibility, oral activity, and central nervous system penetration, suggests that it will be a useful tool compound to define the physiological and pathophysiological roles of NK-3 receptors, in particular in the central nervous system.


Subject(s)
Brain/drug effects , Quinolines/pharmacology , Receptors, Neurokinin-3/antagonists & inhibitors , Animals , Brain/metabolism , CHO Cells , Calcium/metabolism , Cricetinae , Dose-Response Relationship, Drug , Humans , In Vitro Techniques , Iris/drug effects , Iris/physiology , Male , Mice , Mice, Inbred BALB C , Peptide Fragments/pharmacology , Quinolines/pharmacokinetics , Rabbits , Rats , Rats, Sprague-Dawley , Substance P/analogs & derivatives , Substance P/pharmacology
5.
J Biol Chem ; 275(47): 36626-31, 2000 Nov 24.
Article in English | MEDLINE | ID: mdl-10969084

ABSTRACT

Eosinophils have been implicated in the pathogenesis of asthma and other allergic diseases. Several CC chemokines including eotaxin (CCL-11), eotaxin-2 (CCL-24), RANTES (CCL-5), and monocyte chemotactic protein-3 (MCP-3, CCL-7) and 4 (MCP-4, CCL-13) are potent eosinophil chemotactic and activating peptides acting through CC chemokine receptor-3 (CCR3). Thus, antagonism of CCR3 could have a therapeutic role in asthma and other eosinophil-mediated diseases. A high throughput, cellular functional screen was configured using RBL-2H3 cells stably expressing CCR3 (RBL-2H3-CCR3) to identify non-peptide receptor antagonists. A small molecule CCR3 antagonist was identified, SK&F 45523, and chemical optimization led to the generation of a number of highly potent, selective CCR3 antagonists including SB-297006 and SB-328437. These compounds were further characterized in vitro and demonstrated high affinity, competitive inhibition of (125)I-eotaxin and (125)I-MCP-4 binding to human eosinophils. The compounds were potent inhibitors of eotaxin- and MCP-4-induced Ca(2+) mobilization in RBL-2H3-CCR3 cells and eosinophils. Additionally, SB-328437 inhibited eosinophil chemotaxis induced by three ligands that activate CCR3 with similar potencies. Selectivity was affirmed using a panel of 10 seven-transmembrane receptors. This is the first description of a non-peptide CCR3 antagonist, which should be useful in further elucidating the pathophysiological role of CCR3 in allergic inflammatory diseases.


Subject(s)
Benzamides/pharmacology , Cell Movement/drug effects , Chemokines, CC/antagonists & inhibitors , Cytokines/antagonists & inhibitors , Eosinophils/drug effects , Monocyte Chemoattractant Proteins/antagonists & inhibitors , Naphthalenes/pharmacology , Phenylalanine/analogs & derivatives , Receptors, Chemokine/antagonists & inhibitors , Receptors, HIV/antagonists & inhibitors , Asthma/physiopathology , Binding, Competitive , Calcium/metabolism , Cell Line , Chemokine CCL11 , Chemokine CCL24 , Humans , Phenylalanine/pharmacology , Receptors, CCR3 , Receptors, Chemokine/physiology
6.
Mol Pharmacol ; 58(3): 552-9, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10953048

ABSTRACT

There have been proposals that the tachykinin receptor classification should be extended to include a novel receptor, the "neurokinin-4" receptor (NK-4R), which has a close homology with the human NK-3 receptor (hNK-3R). We compared the pharmacological and molecular biological characteristics of the hNK-3R and NK-4R. Binding experiments, with (125)I-[MePhe(7)]-NKB binding to HEK 293 cell membranes transiently expressing the hNK-3R (HEK 293-hNK-3R) or NK-4R (HEK 293-NK-4R), and functional studies (Ca(2+) mobilization in the same cells) revealed a similar profile of sensitivity to tachykinin agonists and antagonists for both receptors; i.e., in binding studies with the hNK-3R, MePhe(7)-NKB > NKB > senktide >> NKA = Substance P; with the NK-4R, MePhe(7)-NKB > NKB = senktide >> Substance P = NKA; and with antagonists, SB 223412 = SR 142801 > SB 222200 >> SR 48968 >> CP 99994 for both hNK-3R and NK-4R. Thus, the pharmacology of the two receptors was nearly identical. However, attempts to isolate or identify the NK-4R gene by using various molecular biological techniques were unsuccessful. Procedures, including nested polymerase chain reaction studies, that used products with restriction endonuclease sites specific for either hNK-3R or NK-4R, failed to demonstrate the presence of NK-4R in genomic DNA from human, monkey, mouse, rat, hamster, or guinea pig, and in cDNA libraries from human lung, brain, or heart, whereas the hNK-3R was detectable in the latter libraries. In view of the failure to demonstrate the presence of the putative NK-4R it is thought to be premature to extend the current tachykinin receptor classification.


Subject(s)
Receptors, Neurokinin-3/metabolism , Receptors, Tachykinin/metabolism , Binding, Competitive , Biological Transport , Calcium/metabolism , Cells, Cultured , DNA, Complementary/analysis , Humans , Polymerase Chain Reaction , Radioligand Assay , Receptors, Neurokinin-3/drug effects , Receptors, Neurokinin-3/genetics , Receptors, Tachykinin/drug effects , Receptors, Tachykinin/genetics , Receptors, Tachykinin/isolation & purification , Restriction Mapping , Tachykinins/metabolism
7.
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
8.
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
9.
Mol Pharmacol ; 57(6): 1190-8, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10825390

ABSTRACT

We describe here the cloning and characterization of a rat homolog of the chemokine receptor CXCR3. The predicted amino acid sequence of rat CXCR3 contains 367 amino acid residues, sharing 96 and 87% amino acid sequence identity to the murine and human CXCR3, respectively. Among a large panel of chemokines tested, only interferon-inducible protein-10 (IP-10), interferon-gamma-induced monokine, and interferon-inducible T cell alpha-chemoattractant demonstrated specific abilities to induce an intracellular calcium mobilization response in human embryonic kidney 293 cells transfected with rat CXCR3 expression vector. (125)I-IP-10 competition binding studies to the CXCR3-transfected human embryonic kidney 293 cells demonstrated that human IP-10 and interferon-inducible T cell alpha-chemoattractant are more potent ligands than human interferon-gamma-induced monokine. Following our previous observation for the induced expression of IP-10 in focal stroke, we demonstrate here the time-dependent up-regulation of CXCR3 mRNA in the rat ischemic cortex after permanent occlusion of the middle cerebral artery. A significant increase in (125)I-IP-10-specific binding to ischemic cerebral cortical samples was obtained and paralleled the increase in CXCR3 mRNA expression. The changes in receptor expression and ligand binding correlate highly with known changes in leukocyte accumulation, and gliosis occurred after focal stroke. These data suggest that CXCR3/IP-10 may be a potential novel therapeutic target in focal stroke. In addition, the cloning of rat CXCR3 provides an important tool for the investigation of the pathophysiological role of CXCR3 in other rodent disease models.


Subject(s)
Chemokines, CXC/metabolism , Receptors, Chemokine/genetics , Stroke/metabolism , Amino Acid Sequence , Animals , Base Sequence , Brain Ischemia/metabolism , Cells, Cultured , Cerebral Arterial Diseases/metabolism , Cerebral Cortex/metabolism , Chemokine CXCL10 , Cloning, Molecular , DNA, Complementary/analysis , Humans , Iodine Radioisotopes , Male , Molecular Sequence Data , RNA, Messenger/biosynthesis , Radioligand Assay , Rats , Rats, Inbred SHR , Receptors, CXCR3 , Receptors, Chemokine/biosynthesis , Sequence Homology, Amino Acid , Transfection
10.
J Biol Chem ; 275(15): 10767-71, 2000 Apr 14.
Article in English | MEDLINE | ID: mdl-10753868

ABSTRACT

Uridine 5'-diphosphoglucose (UDP-glucose) has a well established biochemical role as a glycosyl donor in the enzymatic biosynthesis of carbohydrates. It is less well known that UDP-glucose may possess pharmacological activity, suggesting that a receptor for this molecule may exist. Here, we show that UDP-glucose, and some closely related molecules, potently activate the orphan G protein-coupled receptor KIAA0001 heterologously expressed in yeast or mammalian cells. Nucleotides known to activate P2Y receptors were inactive, indicating the distinctly novel pharmacology of this receptor. The receptor is expressed in a wide variety of human tissues, including many regions of the brain. These data suggest that some sugar-nucleotides may serve important physiological roles as extracellular signaling molecules in addition to their familiar role in intermediary metabolism.


Subject(s)
GTP-Binding Proteins/physiology , Receptors, Cell Surface/physiology , Uridine Diphosphate Glucose/physiology , Humans , Phylogeny , Radioligand Assay , Receptors, Cell Surface/analysis
11.
J Neurosci ; 20(8): 2860-6, 2000 Apr 15.
Article in English | MEDLINE | ID: mdl-10751438

ABSTRACT

PEP-19 is a 6 kDa polypeptide that is highly expressed in select populations of neurons that sometimes demonstrate resistance to degeneration. These include the granule cells of the hippocampus and the Purkinje cells of the cerebellum. Its only identified activity to date is that of binding apo-calmodulin. As a consequence, it has been demonstrated to act as an inhibitor of calmodulin-dependent neuronal nitric oxide synthase in vitro, although PEP-19 regulation of calmodulin-dependent enzymes has never been characterized in intact cells. The activation of the calmodulin-dependent enzyme calmodulin kinase II (CaM kinase II) was studied in PC12 cells that had been transfected so as to express physiological levels of PEP-19. The expression of PEP-19 yielded a stable phenotype that failed to activate CaM kinase II upon depolarization in high K(+). However, CaM kinase II could be fully activated when calcium influx was achieved with ATP. The effect of PEP-19 on CaM kinase II activation was not attributable to changes in the cellular expression of calmodulin. The cellular permeability of the transfected cells to calcium ions also appeared essentially unchanged. The results of this study demonstrated that PEP-19 can regulate CaM kinase II in situ in a manner that was dependent on the stimulus used to mobilize calcium. The selective nature of the regulation by PEP-19 suggests that its function is not to globally suppress calmodulin activity but rather change the manner in which different stimuli can access this activity.


Subject(s)
Calcium Signaling/physiology , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Calmodulin/metabolism , Nerve Tissue Proteins/physiology , Adenosine Triphosphate/pharmacology , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Calcium-Calmodulin-Dependent Protein Kinases/drug effects , Calmodulin-Binding Proteins , Enzyme Activation , Nerve Tissue Proteins/genetics , PC12 Cells , Rats , Transfection
12.
FEBS Lett ; 460(1): 53-6, 1999 Oct 22.
Article in English | MEDLINE | ID: mdl-10571060

ABSTRACT

A cDNA was cloned from a rabbit spleen cDNA library which encoded a G-protein alpha subunit peptide of 374 amino acids, that at the peptide level exhibited 86% and 79% identity with human Galpha16 and mouse G(alpha)15, respectively. The rabbit G(alpha)subunit cDNA was subcloned into a mammalian expression vector and transiently co-transfected into HEK-293 cells along with cDNAs encoding the human C3a, C5a, or nociceptin/orphanin FQ receptors. In all three cases the rabbit G alpha subunit behaved similarly to G(alpha)15 or G(alpha)16 and effectively coupled the transfected receptors to intracellular calcium mobilization pathways. By nucleotide sequence homology and functional activity the rabbit G(alpha) subunit appears to be the ortholog of human G(alpha)16 and mouse G(alpha)15.


Subject(s)
Heterotrimeric GTP-Binding Proteins/genetics , Membrane Proteins , Amino Acid Sequence , Animals , Antigens, CD/genetics , Base Sequence , Calcium/metabolism , Cell Line , Cloning, Molecular , GTP-Binding Protein alpha Subunits, Gq-G11 , Gene Library , Heterotrimeric GTP-Binding Proteins/chemistry , Humans , Mice , Molecular Sequence Data , Rabbits , Receptor, Anaphylatoxin C5a , Receptors, Complement/genetics , Receptors, Opioid/genetics , Sequence Alignment , Spleen/metabolism , Transfection , Nociceptin Receptor
13.
Nature ; 401(6750): 282-6, 1999 Sep 16.
Article in English | MEDLINE | ID: mdl-10499587

ABSTRACT

Urotensin-II (U-II) is a vasoactive 'somatostatin-like' cyclic peptide which was originally isolated from fish spinal cords, and which has recently been cloned from man. Here we describe the identification of an orphan human G-protein-coupled receptor homologous to rat GPR14 and expressed predominantly in cardiovascular tissue, which functions as a U-II receptor. Goby and human U-II bind to recombinant human GPR14 with high affinity, and the binding is functionally coupled to calcium mobilization. Human U-II is found within both vascular and cardiac tissue (including coronary atheroma) and effectively constricts isolated arteries from non-human primates. The potency of vasoconstriction of U-II is an order of magnitude greater than that of endothelin-1, making human U-II the most potent mammalian vasoconstrictor identified so far. In vivo, human U-II markedly increases total peripheral resistance in anaesthetized non-human primates, a response associated with profound cardiac contractile dysfunction. Furthermore, as U-II immunoreactivity is also found within central nervous system and endocrine tissues, it may have additional activities.


Subject(s)
GTP-Binding Proteins/agonists , GTP-Binding Proteins/metabolism , Receptors, Cell Surface/agonists , Receptors, G-Protein-Coupled , Urotensins/pharmacology , Vasoconstrictor Agents/pharmacology , Amino Acid Sequence , Animals , Base Sequence , Calcium/metabolism , Cell Line , Cloning, Molecular , DNA, Complementary , GTP-Binding Proteins/genetics , Humans , Macaca fascicularis , Male , Molecular Sequence Data , Rats , Rats, Sprague-Dawley , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Sequence Homology, Amino Acid , Tissue Distribution , Urotensins/metabolism , Vasoconstrictor Agents/metabolism
14.
Article in English | MEDLINE | ID: mdl-10477044

ABSTRACT

Leukotriene B4 (LTB4) and 12-(R)-hydroxy-5,8,10,14-eicosatetraenoic acid (12-[R]-HETE) have been postulated to contribute to the pathophysiology of inflammatory diseases. SB 201993, (E)-3-[[[[6-(2-carboxyethenyl)-5-[[8-(4-methoxyphenyl)octyl] oxy]-2-pyridinyl] methyl] thio] methyl] benzoic acid, identified from a chemical series designed as ring-fused analogs of LTB4, was evaluated as an antagonist of LTB4- and 12-(R)-HETE-induced responses in vitro and for anti-inflammatory activity in vivo. SB 201993 competitively antagonized [3-H]-LTB4 binding to intact human neutrophils (Ki = 7.6 nM) and to membranes of RBL 2H3 cells expressing the LTB4 receptor (RBL 2H3-LTB4R; IC50 = 154 nM). This compound demonstrated competitive antagonism of LTB4- and 12-(R)-HETE-induced Ca2+ mobilization responses in human neutrophils (IC50s of 131 nM and 105 nM, respectively) and inhibited LTB4-induced Ca2+ mobilization in human cultured keratinocytes (IC50 = 61 nM), RBL 2H3-LTB4R cells (IC50 = 255 nM) and mouse neutrophils (IC50 = 410 nM). SB 201993 showed weak LTD4-receptor binding affinity (Ki = 1.9 microM) and inhibited 5-lipoxygenase (IC50 of 3.6 microM), both in vitro and ex vivo. In vivo, SB 201993 inhibited LTB4-induced neutrophil infiltration in mouse skin and produced dose-related, long lasting topical anti-inflammatory activity against the fluid and cellular phases of arachidonic acid-induced mouse ear inflammation (ED50 of 580 microg/ear and 390 microg/ear, respectively). Similarly, anti-inflammatory activity was also observed in the murine phorbol ester-induced cutaneous inflammation model (ED50 of 770 and 730 microg/ear, respectively, against the fluid and cellular phases). These results indicate that SB 201993 blocks the actions of LTB4 and 12-(R)-HETE and inhibits a variety of inflammatory responses; and thus may be a useful compound to evaluate the role of these mediators in disease models.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Benzoates/pharmacology , Pyridines/pharmacology , Receptors, Leukotriene B4/antagonists & inhibitors , 12-Hydroxy-5,8,10,14-eicosatetraenoic Acid/pharmacology , Animals , Binding, Competitive , Calcimycin/pharmacology , Calcium/blood , Calcium/metabolism , Cell Membrane/metabolism , Cells, Cultured , Guinea Pigs , Humans , Ionophores/pharmacology , Keratinocytes/drug effects , Keratinocytes/metabolism , Leukotriene B4/blood , Leukotriene B4/pharmacology , Male , Mice , Neutrophils/drug effects , Neutrophils/metabolism
15.
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
16.
Nature ; 400(6741): 261-5, 1999 Jul 15.
Article in English | MEDLINE | ID: mdl-10421367

ABSTRACT

The underlying causes of obesity are poorly understood but probably involve complex interactions between many neurotransmitter and neuropeptide systems involved in the regulation of food intake and energy balance. Three pieces of evidence indicate that the neuropeptide melanin-concentrating hormone (MCH) is an important component of this system. First, MCH stimulates feeding when injected directly into rat brains; second, the messenger RNA for the MCH precursor is upregulated in the hypothalamus of genetically obese mice and in fasted animals; and third, mice lacking MCH eat less and are lean. MCH antagonists might, therefore, provide a treatment for obesity. However, the development of such molecules has been hampered because the identity of the MCH receptor has been unknown until now. Here we show that the 353-amino-acid human orphan G-protein-coupled receptor SLC-1 expressed in HEK293 cells binds MCH with sub-nanomolar affinity, and is stimulated by MCH to mobilize intracellular Ca2+ and reduce forskolin-elevated cyclic AMP levels. We also show that SLC-1 messenger RNA and protein is expressed in the ventromedial and dorsomedial nuclei of the hypothalamus, consistent with a role for SLC-1 in mediating the effects of MCH on feeding.


Subject(s)
GTP-Binding Proteins/metabolism , Hypothalamic Hormones/metabolism , Melanins/metabolism , Pituitary Hormones/metabolism , Receptors, Somatostatin/metabolism , Alternative Splicing , Amino Acid Sequence , Animals , Calcium/metabolism , Cell Line , Cloning, Molecular , Cyclic AMP/metabolism , Feeding Behavior , GTP-Binding Proteins/genetics , Humans , Hypothalamus/metabolism , In Situ Hybridization , Ligands , Mice , Molecular Sequence Data , RNA, Messenger/metabolism , Rats , Receptors, Somatostatin/genetics , Recombinant Proteins/metabolism
17.
J Biol Chem ; 274(13): 8367-70, 1999 Mar 26.
Article in English | MEDLINE | ID: mdl-10085065

ABSTRACT

Chimeras were generated between the human anaphylatoxin C3a and C5a receptors (C3aR and C5aR, respectively) to define the structural requirements for ligand binding and discrimination. Chimeric receptors were generated by systematically exchanging between the two receptors four receptor modules (the N terminus, transmembrane regions 1 to 4, the second extracellular loop, and transmembrane region 5 to the C terminus). The mutants were transiently expressed in HEK-293 cells (with or without Galpha-16) and analyzed for cell surface expression, binding of C3a and C5a, and functional responsiveness (calcium mobilization) toward C3a, C5a, and a C3a as well as a C5a analogue peptide. The data indicate that in both anaphylatoxin receptors the transmembrane regions and the second extracellular loop act as a functional unit that is disrupted by any reciprocal exchange. N-terminal substitution confirmed the two-binding site model for the human C5aR, in which the receptor N terminus is required for high affinity binding of the native ligand but not a C5a analogue peptide. In contrast, the human C3a receptor did not require the original N terminus for high affinity binding of and activation by C3a, a result that was confirmed by N-terminal deletion mutants. This indicates a completely different binding mode of the anaphylatoxins to their corresponding receptors. The C5a analogue peptide, but not C5a, was an agonist of the C3aR. Replacement of the C3aR N terminus by the C5aR sequence, however, lead to the generation of a true hybrid C3a/C5a receptor, which bound and functionally responded to both ligands, C3a and C5a.


Subject(s)
Antigens, CD/genetics , Complement C3a/metabolism , Complement C5a/metabolism , Membrane Proteins , Receptors, Complement/genetics , Anaphylatoxins/metabolism , Antigens, CD/metabolism , Binding Sites/genetics , Binding, Competitive/genetics , Calcium/metabolism , Cell Line , Humans , Mutation/genetics , Protein Binding/genetics , Receptor, Anaphylatoxin C5a , Receptors, Complement/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
18.
J Med Chem ; 42(6): 1053-65, 1999 Mar 25.
Article in English | MEDLINE | ID: mdl-10090788

ABSTRACT

Optimization of the previously reported 2-phenyl-4-quinolinecarboxamide NK-3 receptor antagonist 14, with regard to potential metabolic instability of the ester moiety and affinity and selectivity for the human neurokinin-3 (hNK-3) receptor, is described. The ester functionality could be successfully replaced by the ketone (31) or by lower alkyl groups (Et, 21, or n-Pr, 24). Investigation of the substitution pattern of the quinoline ring resulted in the identification of position 3 as a key position to enhance hNK-3 binding affinity and selectivity for the hNK-3 versus the hNK-2 receptor. All of the chemical groups introduced at this position, with the exception of halogens, increased the hNK-3 binding affinity, and compounds 53 (3-OH, SB 223412, hNK-3-CHO binding Ki = 1.4 nM) and 55 (3-NH2, hNK-3-CHO binding Ki = 1.2 nM) were the most potent compounds of this series. Selectivity studies versus the other neurokinin receptors (hNK-2-CHO and hNK-1-CHO) revealed that 53 is about 100-fold selective for the hNK-3 versus hNK-2 receptor, with no affinity for the hNK-1 at concentrations up to 100 microM. In vitro studies demonstrated that 53 is a potent functional antagonist of the hNK-3 receptor (reversal of senktide-induced contractions in rabbit isolated iris sphincter muscles and reversal of NKB-induced Ca2+ mobilization in CHO cells stably expressing the hNK-3 receptor), while in vivo this compound showed oral and intravenous activity in NK-3 receptor-driven models (senktide-induced behavioral responses in mice and senktide-induced miosis in rabbits). Overall, the biological data indicate that (S)-N-(1-phenylpropyl)-3-hydroxy-2-phenylquinoline-4-carboxamide (53, SB 223412) may serve as a pharmacological tool in animal models of disease to assess the functional and pathophysiological role of the NK-3 receptor and to establish therapeutic indications for non-peptide NK-3 receptor antagonists.


Subject(s)
Quinolines/chemical synthesis , Receptors, Neurokinin-3/antagonists & inhibitors , Animals , CHO Cells , Calcium/metabolism , Cell Line , Cloning, Molecular , Cricetinae , Humans , In Vitro Techniques , Iris/drug effects , Iris/physiology , Mice , Miosis/physiopathology , Motor Activity/drug effects , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Muscle, Smooth/physiology , Peptide Fragments/pharmacology , Quinolines/chemistry , Quinolines/metabolism , Rabbits , Radioligand Assay , Receptors, Neurokinin-3/biosynthesis , Structure-Activity Relationship , Substance P/analogs & derivatives , Substance P/pharmacology
19.
J Immunol ; 161(5): 2089-93, 1998 Sep 01.
Article in English | MEDLINE | ID: mdl-9725198

ABSTRACT

The interaction of human anaphylatoxin C4a with the guinea pig (gp) and human (hu) C3a receptors (C3aR) was analyzed using human rC4a, which exhibited C4a-specific activity on guinea pig platelets. A gpC3aR of 475 residues with a large second extracellular loop and a peptide sequence approximately 60% identical to the huC3aR was isolated from a genomic DNA library and found to be expressed in guinea pig heart, lung, and spleen. HEK-293 cells cotransfected with this clone, and a cDNA encoding G alpha-16 specifically bound (Kd = 1.6+/-0.7 nM) and responded functionally to C3a with an intracellular calcium mobilization (ED50 = 0.18+/-0.02 nM). Human rC4a weakly bound to both the hu- and gpC3aR (IC50 > 1 microM). However, only HEK-293 cells expressing the gpC3aR responded functionally to rC4a (ED50 = 8.7+/-0.52 nM), while cells expressing the huC3aR did not (c < or = 1 microM). Thus, through an interaction with the C3aR, huC4a may elicit anaphylatoxic effects in guinea pigs but not in man.


Subject(s)
Complement C4a/agonists , Membrane Proteins , Receptors, Complement/physiology , Amino Acid Sequence , Animals , Base Sequence , Cell Line , Cloning, Molecular , Complement C4a/genetics , Guinea Pigs , Humans , Kidney , Molecular Sequence Data , Platelet Activation/drug effects , Platelet Activation/immunology , Receptors, Complement/chemistry , Receptors, Complement/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacology
20.
J Biol Chem ; 273(17): 10095-8, 1998 Apr 24.
Article in English | MEDLINE | ID: mdl-9553055

ABSTRACT

Interleukin-8 (IL-8) and closely related Glu-Leu-Arg (ELR) containing CXC chemokines, including growth-related oncogene (GRO)alpha, GRObeta, GROgamma, and epithelial cell-derived neutrophil-activating peptide-78 (ENA-78), are potent neutrophil chemotactic and activating peptides, which are proposed to be major mediators of inflammation. IL-8 activates neutrophils by binding to two distinct seven-transmembrane (7-TMR) G-protein coupled receptors CXCR1 (IL-8RA) and CXCR2 (IL-8RB), while GROalpha, GRObeta, GROgamma, and ENA-78 bind to and activate only CXCR2. A chemical lead, which selectively inhibited CXCR2 was discovered by high throughput screening and chemically optimized. SB 225002 (N-(2-hydroxy-4-nitrophenyl)-N'-(2-bromophenyl)urea) is the first reported potent and selective non-peptide inhibitor of a chemokine receptor. It is an antagonist of 125I-IL-8 binding to CXCR2 with an IC50 = 22 nM. SB 225002 showed >150-fold selectivity over CXCR1 and four other 7-TMRs tested. In vitro, SB 225002 potently inhibited human and rabbit neutrophil chemotaxis induced by both IL-8 and GROalpha. In vivo, SB 225002 selectively blocked IL-8-induced neutrophil margination in rabbits. The present findings suggest that CXCR2 is responsible for neutrophil chemotaxis and margination induced by IL-8. This selective antagonist will be a useful tool compound to define the role of CXCR2 in inflammatory diseases where neutrophils play a major role.


Subject(s)
Chemotaxis, Leukocyte/drug effects , Interleukin-8/antagonists & inhibitors , Neutrophils/drug effects , Phenylurea Compounds/pharmacology , Receptors, Chemokine/antagonists & inhibitors , Receptors, Interleukin/antagonists & inhibitors , Animals , CHO Cells , Chemotaxis, Leukocyte/physiology , Cricetinae , Humans , Interleukin-8/physiology , Neutrophils/cytology , Rabbits , Receptors, Interleukin-8B , Recombinant Proteins/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...