Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
J Biol Chem ; 276(31): 28969-75, 2001 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-11387329

RESUMEN

A novel human G protein-coupled receptor named AXOR12, exhibiting 81% homology to the rat orphan receptor GPR54, was cloned from a human brain cDNA library. Heterologous expression of AXOR12 in mammalian cells permitted the identification of three surrogate agonist peptides, all with a common C-terminal amidated motif. High potency agonism, indicative of a cognate ligand, was evident from peptides derived from the gene KiSS-1, the expression of which prevents metastasis in melanoma cells. Quantitative reverse transcriptase-polymerase chain reaction was used to study the expression of AXOR12 and KiSS-1 in a variety of tissues. The highest levels of expression of AXOR12 mRNA were observed in brain, pituitary gland, and placenta. The highest levels of KiSS-1 gene expression were observed in placenta and brain. A polyclonal antibody raised to the C terminus of AXOR12 was generated and used to show localization of the receptor to neurons in the cerebellum, cerebral cortex, and brainstem. The biological significance of these expression patterns and the nature of the putative cognate ligand for AXOR12 are discussed.


Asunto(s)
Proteínas de Unión al GTP Heterotriméricas/metabolismo , Proteínas/genética , Proteínas/metabolismo , Receptores de Superficie Celular/genética , Secuencia de Aminoácidos , Animales , Encéfalo/metabolismo , Células CHO , Cricetinae , Femenino , Genes Supresores de Tumor , Humanos , Cinética , Kisspeptinas , Ligandos , Melanoma/genética , Datos de Secuencia Molecular , Nephropidae , Neuronas/metabolismo , Especificidad de Órganos , Fragmentos de Péptidos/farmacología , Hipófisis/metabolismo , Placenta/metabolismo , Embarazo , Proteínas/química , Ratas , Receptores de Superficie Celular/química , Receptores Acoplados a Proteínas G , Receptores de Kisspeptina-1 , Receptores de Neuropéptido/química , Receptores de Neuropéptido/genética , Proteínas Recombinantes/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Anémonas de Mar , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Transfección , Proteínas Supresoras de Tumor
2.
Eur J Pharmacol ; 413(2-3): 143-50, 2001 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-11226387

RESUMEN

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.


Asunto(s)
Calcio/metabolismo , Receptores de Neuroquinina-3/fisiología , Secuencia de Aminoácidos/fisiología , Animales , Clonación Molecular/métodos , Humanos , Ratones , Datos de Secuencia Molecular , Neuroquinina A/metabolismo , Neuroquinina A/farmacología , Quinolinas/química , Quinolinas/metabolismo , Quinolinas/farmacología , Receptores de Neuroquinina-3/efectos de los fármacos
3.
Br J Pharmacol ; 131(6): 1050-4, 2000 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11082110

RESUMEN

1. The aim of this study was to characterize the pharmacological profile of the GABA(B1)/GABA(B2) heterodimeric receptor expressed in Chinese hamster ovary (CHO) cells. We have compared receptor binding affinity and functional activity for a series of agonists and antagonists. 2. The chimeric G-protein, G(qi5), was used to couple receptor activation to increases in intracellular calcium for functional studies on the Fluorimetric Imaging Plate Reader (FLIPR), using a stable GABA(B1)/GABA(B2)/G(qi5) CHO cell line. [(3)H]-CGP-54626 was used in radioligand binding studies in membranes prepared from the same cell line. 3. The pharmacological profile of the recombinant GABA(B1/B2) receptor was consistent with that of native GABA(B) receptors in that it was activated by GABA and baclofen and inhibited by CGP-54626A and SCH 50911. 4. Unlike native receptors, the GABA(B1)/GABA(B2)/G(qi5) response was not inhibited by high microMolar concentration of phaclofen, saclofen or CGP 35348. 5. This raises the possibility that the GABA(B1)/GABA(B2)/G(qi5) recombinant receptor may represent the previously described GABA(B) receptor subtype which is relatively resistant to inhibition by phaclofen.


Asunto(s)
Baclofeno/análogos & derivados , Agonistas del GABA/metabolismo , Antagonistas del GABA/metabolismo , Receptores de GABA-B/metabolismo , Receptores de GABA/metabolismo , Animales , Baclofeno/metabolismo , Baclofeno/farmacología , Células CHO , Cricetinae , Relación Dosis-Respuesta a Droga , Antagonistas del GABA/farmacología , Humanos , Ácido gamma-Aminobutírico/metabolismo
4.
Biochem Biophys Res Commun ; 273(3): 805-10, 2000 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-10891327

RESUMEN

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.


Asunto(s)
Proteínas de Unión al GTP/metabolismo , Lisofosfolípidos/metabolismo , Receptores de Superficie Celular/genética , Receptores Acoplados a Proteínas G , Secuencia de Aminoácidos , Secuencia de Bases , Calcio/metabolismo , Mapeo Cromosómico , Cromosomas Humanos Par 1 , Clonación Molecular , Activación Enzimática , Humanos , Células Jurkat , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Datos de Secuencia Molecular , Fosforilación , Receptores de Superficie Celular/química , Receptores de Superficie Celular/metabolismo , Receptores del Ácido Lisofosfatídico , Homología de Secuencia de Aminoácido , Células Tumorales Cultivadas
5.
J Biol Chem ; 275(34): 25965-71, 2000 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-10851242

RESUMEN

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.


Asunto(s)
Neuropéptidos/metabolismo , Oligopéptidos/metabolismo , Receptores de Neuropéptido/metabolismo , Secuencia de Aminoácidos , Arrestinas/metabolismo , Secuencia de Bases , Calcio/metabolismo , Línea Celular , FMRFamida/farmacología , Humanos , Ligandos , Datos de Secuencia Molecular , Receptores de Neuropéptido/genética , beta-Arrestinas
6.
J Biol Chem ; 275(27): 20247-50, 2000 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-10811630

RESUMEN

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.


Asunto(s)
Proteínas de Unión al GTP/metabolismo , Proteínas de la Membrana , Neuropéptidos/farmacología , Receptores de Superficie Celular/agonistas , Receptores de Neurotransmisores , Calcio/metabolismo , Línea Celular , Clonación Molecular , Regulación de la Expresión Génica , Humanos , Fosfatos de Inositol/metabolismo , Neuropéptidos/genética , ARN Mensajero/análisis , ARN Mensajero/metabolismo , Receptores de Superficie Celular/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección
7.
Eur J Biochem ; 267(3): 919-26, 2000 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-10651831

RESUMEN

Murine MARCO has been identified recently in subsets of macrophages located in the peritoneum, marginal zone of the spleen, and the medullary cord of lymph nodes, where it has been proposed that it serves as a bacteria-binding receptor. A scavenger receptor family member with an extended collagenous domain, murine MARCO has also been demonstrated in atherosclerotic lesions of susceptible mice. We report here the identification, tissue and chromosomal localization, and pharmacological characterization of human (h)MARCO. hMARCO was identified from a macrophage cDNA library by electronic screening with the murine MARCO sequence. Nucleotide sequence analysis confirmed that the full-length hMARCO clone encoded a 519-amino acid protein sharing 68.5% identity with murine MARCO. RNA blot analysis indicated that the hMARCO transcript is 2.0 kb in length and is predominantly expressed in human lung, liver, and lymph nodes. Radiation hybrid mapping localized hMARCO to chromosome 2q14. Ligand-binding studies of COS cells expressing hMARCO demonstrated significant specific binding of both Escherichia coli and Staphylococcus aureus. In contrast, the hMARCO receptor expressed in COS cells did not specifically bind the scavenger receptor ligand acetylated low-density lipoprotein (LDL), despite its similarity to the elongated collagen-like binding domain of the macrophage scavenger receptor. In addition, acetylated (Ac)LDL and oxidized (Ox)LDL did not inhibit E. coli binding to hMARCO. These data suggest that hMARCO may play an important role in host defense, but it has no obvious role in the accumulation of modified lipoproteins during atherogenesis.


Asunto(s)
Macrófagos/metabolismo , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Secuencia de Aminoácidos , Animales , Adhesión Bacteriana , Secuencia de Bases , Células COS , Mapeo Cromosómico , Cromosomas Humanos Par 2/genética , Clonación Molecular , Cartilla de ADN/genética , Escherichia coli/inmunología , Femenino , Humanos , Macrófagos/inmunología , Macrófagos/microbiología , Masculino , Ratones , Datos de Secuencia Molecular , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homología de Secuencia de Aminoácido , Especificidad de la Especie , Distribución Tisular
8.
J Pharmacol Exp Ther ; 292(1): 247-53, 2000 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-10604954

RESUMEN

Screening of porcine cerebellum cDNA library with porcine endothelin(B) (ET(B)) receptor cDNA revealed a novel ET(B) receptor cDNA that is distinctly different from the wild-type ET(B) receptor in length and the amino acid sequence at the C-terminal end. This sequence appears to represent alternate splicing of the carboxy terminal end of ET(B) receptor, resulting in a polypeptide of 429 amino acids in length, which is 14 amino acids shorter than the wild-type porcine ET(B) receptor. Characterization of the wild-type and alternately spliced ET(B) receptors expressed in COS cells revealed that both receptors displayed very similar binding [apparent dissociation constant (K(d)) and maximum binding (B(max)) for (125)I-ET-1 were 71 pM and 1.6 pmol/mg protein for wild-type and 81 pM and 1.2 pmol/mg protein for splice variant ET(B) receptors] as well as functional properties. These data suggest that the differences in the amino acids at the C-terminal end had no effect on binding or functional coupling of these alternately spliced ET(B) receptors.


Asunto(s)
Células COS/metabolismo , Cerebelo/química , Receptores de Endotelina/química , Empalme Alternativo , Secuencia de Aminoácidos , Animales , Northern Blotting , Relación Dosis-Respuesta a Droga , Biblioteca de Genes , Técnicas In Vitro , Datos de Secuencia Molecular , Forbol 12,13-Dibutirato/farmacología , Fosfatidilinositoles/metabolismo , Ensayo de Unión Radioligante , Receptor de Endotelina B , Receptores de Endotelina/clasificación , Porcinos
9.
Nature ; 401(6750): 282-6, 1999 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-10499587

RESUMEN

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.


Asunto(s)
Proteínas de Unión al GTP/agonistas , Proteínas de Unión al GTP/metabolismo , Receptores de Superficie Celular/agonistas , Receptores Acoplados a Proteínas G , Urotensinas/farmacología , Vasoconstrictores/farmacología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Calcio/metabolismo , Línea Celular , Clonación Molecular , ADN Complementario , Proteínas de Unión al GTP/genética , Humanos , Macaca fascicularis , Masculino , Datos de Secuencia Molecular , Ratas , Ratas Sprague-Dawley , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Homología de Secuencia de Aminoácido , Distribución Tisular , Urotensinas/metabolismo , Vasoconstrictores/metabolismo
10.
Br J Pharmacol ; 128(1): 1-3, 1999 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10498827

RESUMEN

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.


Asunto(s)
Calcio/metabolismo , Proteínas Portadoras/farmacología , Péptidos y Proteínas de Señalización Intracelular , Neuropéptidos/farmacología , Receptores de Neuropéptido/metabolismo , Compuestos de Anilina , Animales , Células CHO , Calcio/antagonistas & inhibidores , Calcio/farmacología , Señalización del Calcio/efectos de los fármacos , Proteínas Portadoras/antagonistas & inhibidores , Cricetinae , Relación Dosis-Respuesta a Droga , Colorantes Fluorescentes , Humanos , Neuropéptidos/antagonistas & inhibidores , Receptores de Orexina , Orexinas , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosfolipasa D/antagonistas & inhibidores , Fosfolipasa D/metabolismo , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/metabolismo , Receptores Acoplados a Proteínas G , Receptores de Neuropéptido/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Tapsigargina/farmacología , Factores de Tiempo , Fosfolipasas de Tipo C/antagonistas & inhibidores , Fosfolipasas de Tipo C/metabolismo , Xantenos
11.
Mol Pharmacol ; 56(3): 657-63, 1999 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10462554

RESUMEN

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.


Asunto(s)
Proteínas de la Membrana , Receptores de Leucotrienos/genética , Secuencia de Aminoácidos , Secuencia de Bases , Sitios de Unión , Transporte Biológico/efectos de los fármacos , Calcio/metabolismo , Células Cultivadas , Clonación Molecular , Humanos , Leucotrieno D4/farmacología , Datos de Secuencia Molecular , Toxina del Pertussis , Receptores de Leucotrienos/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Virulencia de Bordetella/farmacología
12.
J Pharmacol Exp Ther ; 289(3): 1277-85, 1999 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10336517

RESUMEN

Class A scavenger receptor (SR-A) antagonists may prevent the initiation of atherosclerosis, because a recent report found that SR-A/apolipoprotein E (apoE) double-knockout mice had 60% smaller lesions than apoE single-knockout littermates. We transfected human embryonic kidney (HEK) 293 cells with SR-A type I or II receptors to find small-molecule antagonists. Uptake of 1,1'-dioctadecyl-3,3,3', 3'-tetramethylindocarbocyanine perchlorate-labeled acetylated low-density lipoprotein (DiI-AcLDL) showed that among common polyanionic ligands, polyinosine was the most potent, with an IC50 of 0.74 microgram/ml, whereas the novel compound (E)-methyl 4-chloro-alpha-[4-(4-chlorophenyl)-1, 5-dihydro-3-hydroxy-5-oxo-1-(2-thiazolyl)-2H-pyrrol-2-ylidene]benzene acetate gave an IC50 of 6.1 microgram/ml (13 microM). The novel antagonist also inhibited DiI-AcLDL uptake in cultured human peripheral and rat peritoneal macrophages with IC50 values of 21 microM and 17 microM, respectively. With [125I]AcLDL as ligand for transfected HEK 293 cells, binding/uptake and degradation at 37 degrees C for 5 h was saturable and selective. In a comparison of both types of receptor, we found no difference between the capacity of SR-AI or SR-AII for either binding or degradation. Polyinosine competed both [125I]AcLDL binding and degradation with a Ki of 1 microgram/ml, whereas the novel antagonist competed with a Ki of 19 microgram/ml (40 microM) and 8.6 microgram/ml (18 microM), respectively, for binding and degradation. Saturation binding in the presence of the ionophore monensin indicated that the novel compound behaved as a noncompetitive antagonist and perhaps as an allosteric effector. This is the first report to describe a small-molecule macrophage scavenger receptor antagonist. Utilization of this permanently transfected HEK 293 cell line will allow the identification of more potent macrophage scavenger receptor antagonists, so that their utility as therapeutics for atherosclerosis can be determined.


Asunto(s)
Macrófagos Peritoneales/fisiología , Macrófagos/fisiología , Receptores Inmunológicos/antagonistas & inhibidores , Animales , Unión Competitiva , Transporte Biológico/efectos de los fármacos , Línea Celular , Células Cultivadas , Clonación Molecular , Sulfato de Dextran/farmacología , Humanos , Cinética , Ligandos , Lipoproteínas LDL/farmacocinética , Macrófagos/efectos de los fármacos , Macrófagos Peritoneales/efectos de los fármacos , Ratones , Ratones Noqueados , Fenilacetatos/farmacología , Poli I/farmacología , Ratas , Receptores Inmunológicos/genética , Receptores Inmunológicos/fisiología , Receptores Depuradores , Proteínas Recombinantes/antagonistas & inhibidores , Receptores Depuradores de Clase A , Tiazoles/farmacología , Transfección
14.
Endocrinology ; 139(4): 1678-83, 1998 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-9528949

RESUMEN

Calcitonin gene-related peptide (CGRP) receptors (CGRP-Rs) are widely distributed throughout the central and peripheral nervous systems. A novel CGRP-R was identified from a porcine lung complementary DNA library. Sequence analysis indicated that the CGRP-R is 462 amino acids in length and shares 93% sequence identity with the human CGRP-R. Northern blot analysis indicated a messenger RNA species of 5.4 kilobases, which is abundantly expressed in the lung. Ligand binding studies of the cloned CGRP-R expressed in human embryonic kidney (HEK-293) cells showed the presence of high affinity receptor for CGRP with a Kd of 38.5 pM. The pharmacological profiles of various ligands competing for [125I]CGRP binding to the expressed receptor were in accordance with those for the natural receptor. Binding of [125I]CGRP to the expressed receptor was decreased in the presence of a nonhydrolyzable analog of GTP, guanosine 5' (gamma-thio)-triphosphate. In functional studies, CGRP stimulated the activation of adenylyl cyclase with an EC50 of 2.5 nM. The linear analog of CGRP, diacetoamidomethyl cysteine CGRP, did not affect adenylyl cyclase activity on its own or in the presence of CGRP. Furthermore, the CGRP receptor antagonists, CGRP-(8-37)alpha, inhibited the CGRP-mediated response in a competitive manner. Collectively, the binding and functional data demonstrate that we have cloned a porcine CGRP type 1 receptor. The availability of the CGRP-R complementary DNA will allow us to examine its participation in pathophysiological processes.


Asunto(s)
Clonación Molecular , Receptores de Péptido Relacionado con el Gen de Calcitonina/genética , Porcinos/genética , Secuencia de Aminoácidos , Animales , Unión Competitiva , Northern Blotting , Péptido Relacionado con Gen de Calcitonina/metabolismo , Péptido Relacionado con Gen de Calcitonina/farmacología , Línea Celular , ADN Complementario/genética , Embrión de Mamíferos , Proteínas de Unión al GTP/metabolismo , Humanos , Riñón , Pulmón/química , Datos de Secuencia Molecular , ARN Mensajero/análisis , Receptores de Péptido Relacionado con el Gen de Calcitonina/química , Receptores de Péptido Relacionado con el Gen de Calcitonina/metabolismo , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Análisis de Secuencia
15.
Cell ; 92(4): 573-85, 1998 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-9491897

RESUMEN

The hypothalamus plays a central role in the integrated control of feeding and energy homeostasis. We have identified two novel neuropeptides, both derived from the same precursor by proteolytic processing, that bind and activate two closely related (previously) orphan G protein-coupled receptors. These peptides, termed orexin-A and -B, have no significant structural similarities to known families of regulatory peptides. prepro-orexin mRNA and immunoreactive orexin-A are localized in neurons within and around the lateral and posterior hypothalamus in the adult rat brain. When administered centrally to rats, these peptides stimulate food consumption. prepro-orexin mRNA level is up-regulated upon fasting, suggesting a physiological role for the peptides as mediators in the central feedback mechanism that regulates feeding behavior.


Asunto(s)
Proteínas Portadoras/genética , Conducta Alimentaria/fisiología , Proteínas de Unión al GTP/genética , Hipotálamo/química , Péptidos y Proteínas de Señalización Intracelular , Neuropéptidos/genética , Receptores de Neuropéptido/genética , Animales , Células CHO , Proteínas Portadoras/aislamiento & purificación , Proteínas Portadoras/farmacología , Cromatografía Líquida de Alta Presión , Cricetinae , Ayuno/fisiología , Humanos , Hipotálamo/citología , Riñón/citología , Masculino , Datos de Secuencia Molecular , Neuronas/química , Neuronas/efectos de los fármacos , Neuropéptidos/aislamiento & purificación , Neuropéptidos/farmacología , Receptores de Orexina , Orexinas , Precursores de Proteínas/genética , Precursores de Proteínas/aislamiento & purificación , ARN Mensajero/metabolismo , Conejos , Ratas , Ratas Wistar , Receptores Acoplados a Proteínas G , Receptores de Neuropéptido/química , Receptores de Neuropéptido/aislamiento & purificación , Homología de Secuencia de Aminoácido
16.
J Leukoc Biol ; 62(5): 667-75, 1997 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-9365122

RESUMEN

Eotaxin has been found to bind exclusively to a single chemokine receptor, CCR3. Using expression sequence tag screening of an activated monocyte library, a second chemokine has been identified; it was expressed and purified from a Drosophila cell culture system and appears to only activate CCR3. Eotaxin-2, MPIF-2, or CKbeta-6, is a human CC chemokine with low amino acid sequence identity to other chemokines. Eotaxin-2 promotes chemotaxis and Ca2+ mobilization in human eosinophils but not in neutrophils or monocytes. Cross-desensitization calcium mobilization experiments using purified eosinophils indicate that eotaxin and MCP-4, but not RANTES, MIP-1alpha, or MCP-3, can completely cross-desensitize the calcium response to eotaxin-2 on these cells, indicating that eotaxin-2 shares the same receptor used by eotaxin and MCP-4. Eotaxin-2 was the most potent eosinophil chemoattractant of all the chemokines tested. Eotaxin-2 also displaced 125I-eotaxin bound to the cloned CCR3 stably expressed in CHO cells (CHO-CCR3) and to freshly isolated human eosinophils with affinities similar to eotaxin and MCP-4. 125I-Eotaxin-2 binds with high affinity to eosinophils and both eotaxin and cold eotaxin-2 displace the ligand with equal affinity. Eotaxin and eotaxin-2 promote a Ca2+ transient in RBL-2H3 cells stably transfected with CCR3 (RBL-2H3-CCR3) and both ligands cross-desensitized the response of the other but not the response to LTD4. The data indicate that eotaxin-2 is a potent eosinophil chemotactic chemokine exerting its activity solely through the CCR3 receptor.


Asunto(s)
Quimiocinas CC , Quimiocinas/fisiología , Eosinófilos/fisiología , Receptores de Quimiocina/metabolismo , Secuencia de Aminoácidos , Animales , Unión Competitiva , Células CHO/metabolismo , Calcio/metabolismo , Movimiento Celular/fisiología , Quimiocina CCL11 , Quimiocina CCL24 , Quimiocina CCL8 , Quimiocinas/genética , Quimiocinas/aislamiento & purificación , Clonación Molecular , Cricetinae , Citocinas/genética , ADN Complementario/genética , Eosinófilos/efectos de los fármacos , Eosinófilos/metabolismo , Humanos , Datos de Secuencia Molecular , Proteínas Quimioatrayentes de Monocitos/genética , Ratas , Receptores CCR3 , Receptores de Quimiocina/fisiología
17.
J Pharmacol Exp Ther ; 281(3): 1303-11, 1997 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-9190866

RESUMEN

The in vitro and in vivo pharmacological profile of SB 223412 [(S)-(-)-N-(alpha-ethylbenzyl)-3-hydroxy-2-phenylquinoline-4-carbo xamide], a novel human NK-3 (hNK-3) receptor antagonist, is described. SB 223412 demonstrated enantioselective affinity for inhibition of [125I][MePhe7]neurokinin B (NKB) binding to membranes of CHO cells expressing the hNK-3 receptor (CHO hNK-3). SB 223412, the (S)-isomer, (Ki = 1.0 nM), has similar affinity as the natural ligand, NKB (Ki = 0.8 nM) and another nonpeptide NK-3 receptor antagonist, SR 142801 (Ki = 1.2 nM). SB 223412 was selective for hNK-3 receptors compared with hNK-1 (>10,000-fold selective) and hNK-2 receptors (>140-fold selective), and selectivity was further demonstrated by its lack of effect, in concentrations up to 1 or 10 microM, in >60 receptor, enzyme and ion channel assays. SB 223412 enantioselectively inhibited the NKB-induced Ca++ mobilization in HEK 293 cells stably expressing the hNK-3 receptor. SB 223412 (10-1,000 nM) produced concentration-dependent rightward shifts in NKB-induced Ca++ mobilization concentration-response curves with a Kb value of 3 nM. In addition, SB 223412 antagonized senktide-induced contraction in the isolated rabbit iris sphincter muscle (Kb = 1.6 nM). In mice, oral administration of SB 223412 produced dose-dependent inhibition of behavioral responses induced by the NK-3 receptor-selective agonist, senktide (ED50 = 12.2 mg/kg). Pharmacokinetic evaluation of SB 223412 in rat and dog indicated low plasma clearance, oral bioavailability and high and sustained plasma concentrations after 4 to 8 mg/kg oral dosages. The preclinical profile of SB 223412 (high affinity, selectivity, reversibility and oral activity) suggests that it will be a useful tool compound to define the physiological and pathophysiological roles of NK-3 receptors.


Asunto(s)
Piperidinas/farmacología , Quinolinas/farmacología , Receptores de Neuroquinina-3/efectos de los fármacos , Receptores de Taquicininas/antagonistas & inhibidores , Animales , Células CHO/efectos de los fármacos , Calcio/metabolismo , Cricetinae , Perros , Humanos , Ratones , Piperidinas/farmacocinética , Conejos , Ensayo de Unión Radioligante , Ratas
18.
J Biol Chem ; 272(26): 16404-13, 1997 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-9195948

RESUMEN

Here we describe the characterization of a novel human CC chemokine, tentatively named monocyte chemotactic protein (MCP-4). This chemokine was detected by random sequencing of expressed sequence tags in cDNA libraries. The full-length cDNA revealed an open reading frame for a 98-amino acid residue protein, and a sequence alignment with known CC chemokines showed high levels of similarity (59-62%) with MCP-1, MCP-3, and eotaxin. MCP-4 cDNA was cloned into Drosophila S2 cells, and the mature protein (residues 24-98) was purified from the conditioned medium. Recombinant MCP-4 induced a potent chemotactic response (EC50 = 2.88 +/- 0.15 nM) and a transient rise in cytosolic calcium concentration in fresh human peripheral blood monocytes but not in neutrophils. Binding studies in monocytes showed that MCP-4 and MCP-3 were very potent in displacing high affinity binding of 125I-MCP-1 (IC50 for MCP-4, MCP-3, and unlabeled MCP-1 of 2.1 +/- 1.4, 0.85-1.6, and 0.7 +/- 0.2 nM respectively), suggesting that all three chemokines interact with the CC chemokine receptor-2 (MCP-1 receptor). This was confirmed in binding studies with Chinese hamster ovary cells, stably transfected with the CC chemokine 2B receptor. Northern blot analysis in extracts of normal human tissues showed expression of mRNA for MCP-4 in small intestine, thymus, and colon, but the level of protein expression was too low to be detected in Western blot analysis. However, expression of MCP-4 protein was demonstrated by immunohistochemistry in human atherosclerotic lesion and found to be associated with endothelial cells and macrophages.


Asunto(s)
Proteínas Quimioatrayentes de Monocitos/metabolismo , Receptores de Quimiocina , Receptores de Citocinas/metabolismo , Secuencia de Aminoácidos , Animales , Arteriosclerosis/metabolismo , Unión Competitiva , Western Blotting , Células CHO , Calcio/metabolismo , Clonación Molecular , Cricetinae , Humanos , Datos de Secuencia Molecular , Proteínas Quimioatrayentes de Monocitos/análisis , Proteínas Quimioatrayentes de Monocitos/farmacología , ARN Mensajero/análisis , Receptores CCR2 , Proteínas Recombinantes/biosíntesis
19.
J Clin Invest ; 98(10): 2381-7, 1996 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-8941657

RESUMEN

Chronic metabolic acidosis increases proximal tubular citrate uptake and metabolism. The present study addressed the effect of chronic metabolic acidosis on a cytosolic enzyme of citrate metabolism, ATP citrate lyase. Chronic metabolic acidosis caused hypocitraturia in rats and increased renal cortical ATP citrate lyase activity by 67% after 7 d. Renal cortical ATP citrate lyase protein abundance increased by 29% after 3 d and by 141% after 7 d of acid diet. No significant change in mRNA abundance could be detected. Hypokalemia, which causes only intracellular acidosis, caused hypocitraturia and increased renal cortical ATP citrate lyase activity by 28%. Conversely, the hypercitraturia of chronic alkali feeding was associated with no change in ATP citrate lyase activity. Inhibition of ATP citrate lyase with the competitive inhibitor, 4S-hydroxycitrate, significantly abated hypocitraturia and increased urinary citrate excretion fourfold in chronic metabolic acidosis and threefold in K+-depletion. In summary, the hypocitraturia of chronic metabolic acidosis is associated with an increase in ATP citrate lyase activity and protein abundance, and is partly reversed by inhibition of this enzyme. These results suggest an important role for ATP citrate lyase in proximal tubular citrate metabolism.


Asunto(s)
ATP Citrato (pro-S)-Liasa/fisiología , Citratos/metabolismo , Riñón/metabolismo , ATP Citrato (pro-S)-Liasa/antagonistas & inhibidores , ATP Citrato (pro-S)-Liasa/genética , ATP Citrato (pro-S)-Liasa/inmunología , Acidosis Tubular Renal/metabolismo , Animales , Northern Blotting , Citratos/farmacología , Alimentos Formulados , Hipopotasemia/metabolismo , Immunoblotting , Corteza Renal/metabolismo , Masculino , ARN Mensajero/biosíntesis , Ratas , Ratas Sprague-Dawley , Cálculos Urinarios/metabolismo
20.
J Biol Chem ; 271(41): 25300-7, 1996 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-8810293

RESUMEN

Endothelin receptors are widely distributed throughout a number of tissues. A novel ETB receptor splice variant (ETB-SVR) was identified from a human placental cDNA library. Sequence analysis indicated that the ETB-SVR is 436 amino acids long and shares 91% identity to the human ETB-R. Northern blot analysis indicated an mRNA species of 2.7 kilobases, which is expressed in the lung, placenta, kidney, and skeletal muscle. Ligand binding studies of the cloned ETB-SVR and ETB-R receptors expressed in COS cells showed that ET peptides exhibited similar potency in displacing 125I-ET-1 binding. Functional studies showed that ET-1, ET-3, and sarafotoxin 6c displayed similar potencies for inositol phosphates accumulation in ETB-R-transfected COS cells, whereas no increase in inositol phosphate accumulation was observed in ETB-SVR-transfected cells. In addition, exposure of ETB-R-transfected cells to ET-1 caused an increase in the intracellular acidification rate whereas ETB-SVR-transfected cells did not respond to ET-1. These data suggest that the ETB-SVR and ETB-R are functionally distinct and the difference in the amino acid sequences between the two receptors may determine functional coupling. Availability of cDNA clones for endothelin receptors can facilitate our understanding of the role of ET in the pathophysiology of various diseases.


Asunto(s)
Empalme Alternativo , Variación Genética , Estructura Secundaria de Proteína , Receptores de Endotelina/biosíntesis , Receptores de Endotelina/química , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Unión Competitiva , Células COS , Chlorocebus aethiops , Cartilla de ADN , Endotelina-1/farmacología , Endotelina-3/metabolismo , Endotelina-3/farmacología , Endotelinas/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Inositol/metabolismo , Cinética , Modelos Estructurales , Datos de Secuencia Molecular , Especificidad de Órganos , Fragmentos de Péptidos/metabolismo , Fosfatidilinositoles/metabolismo , Receptor de Endotelina B , Receptores de Endotelina/metabolismo , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Homología de Secuencia de Aminoácido , Porcinos , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA