Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
1.
Clin Pharmacol Ther ; 84(4): 475-80, 2008 Oct.
Article in English | MEDLINE | ID: mdl-19238652

ABSTRACT

A number of issues have remained unanswered in the design of "thorough QT"(TQT) studies. In this randomized, placebo-controlled, two-period crossover study in 20 healthy subjects, replicate electrocardiograms (ECGs) were recorded on a digital 12-lead Holter recorder, extracted in a core ECG laboratory, and interpreted manually by a cardiologist. The observed within-subject variability was slightly greater when time-matched baselines were employed than when predose baselines were employed, whereas the magnitude of the increase in QTc was similar for both. Moxifloxacin 400 mg was associated with an observed 7.5-12.5 ms increase in the mean placebo- and baseline-corrected QTc interval. A PK-QTc model estimated a 3.9 ms increase in the QTc interval for every 1,000 ng/ml increase in moxifloxacin concentration. The QTc increases associated with moxifloxacin support the appropriateness of its use as a positive control in TQT studies. This crossover study failed to justify the use of time-matched baselines rather than the less resource-intensive predose definition of baseline.


Subject(s)
Anti-Infective Agents/adverse effects , Aza Compounds/adverse effects , Long QT Syndrome/chemically induced , Quinolines/adverse effects , Research Design , Adult , Anti-Infective Agents/administration & dosage , Aza Compounds/administration & dosage , Cross-Over Studies , Dose-Response Relationship, Drug , Electrocardiography , Female , Fluoroquinolones , Humans , Long QT Syndrome/physiopathology , Male , Moxifloxacin , Pilot Projects , Quinolines/administration & dosage
2.
Int J Clin Pharmacol Ther ; 44(2): 83-92, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16502768

ABSTRACT

OBJECTIVE: To assess the bioequivalence of an ezetimibe/simvastatin (EZE/SIMVA) combination tablet compared to the coadministration of ezetimibe and simvastatin as separate tablets (EZE + SIMVA). METHODS: In this open-label, randomized, 2-part, 2-period crossover study, 96 healthy subjects were randomly assigned to participate in each part of the study (Part I or II), with each part consisting of 2 single-dose treatment periods separated by a 14-day washout. Part I consisted of Treatments A (EZE 10 mg + SIMVA 10 mg) and B (EZE/SIMVA 10/10 mg/mg) and Part II consisted of Treatments C (EZE 10 mg + SIMVA 80 mg) and D (EZE/SIMVA 10/80 mg/mg). Blood samples were collected up to 96 hours post-dose for determination of ezetimibe, total ezetimibe (ezetimibe + ezetimibe glucuronide), simvastatin and simvastatin acid (the most prevalent active metabolite of simvastatin) concentrations. Ezetimibe and simvastatin acid AUC(0-last) were predefined as primary endpoints and ezetimibe and simvastatin acid Cmax were secondary endpoints. Bioequivalence was achieved if 90% confidence intervals (CI) for the geometric mean ratios (GMR) (single tablet/coadministration) of AUC(0-last) and Cmax fell within prespecified bounds of (0.80, 1.25). RESULTS: The GMRs of the AUC(0-last) and Cmax for ezetimibe and simvastatin acid fell within the bioequivalence limits (0.80, 1.25). EZE/ SIMVA and EZE + SIMVA were generally well tolerated. CONCLUSIONS: The lowest and highest dosage strengths of EZE/SIMVA tablet were bioequivalent to the individual drug components administered together. Given the exact weight multiples of the EZE/SIMVA tablet and linear pharmacokinetics of simvastatin across the marketed dose range, bioequivalence of the intermediate tablet strengths (EZE/SIMVA 10/20 mg/mg and EZE/SIMVA 10/40 mg/mg) was inferred, although these dosages were not tested directly. These results indicate that the safety and efficacy profile of EZE + SIMVA coadministration therapy can be applied to treatment with the EZE/SIMVA tablet across the clinical dose range.


Subject(s)
Anticholesteremic Agents/pharmacokinetics , Azetidines/pharmacokinetics , Simvastatin/pharmacokinetics , Adolescent , Adult , Analysis of Variance , Anticholesteremic Agents/administration & dosage , Anticholesteremic Agents/adverse effects , Area Under Curve , Azetidines/administration & dosage , Azetidines/adverse effects , Cross-Over Studies , Drug Combinations , Drug Therapy, Combination , Ezetimibe , Female , Humans , Male , Middle Aged , Reference Values , Simvastatin/administration & dosage , Simvastatin/adverse effects , Tablets , Therapeutic Equivalency , Time Factors , Treatment Outcome
3.
Am J Physiol Heart Circ Physiol ; 284(4): H1429-40, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12505877

ABSTRACT

IGF-I and IGF-II are single-chain polypeptide growth factors that regulate pleiotropic cellular responses. We have characterized the effect of recombinant IGF proteins, as well as third-generation adenoviral vectors encoding either IGF-I or IGF-II genes, on cardiomyocyte apoptosis and on angiogenesis. We found that endothelial cells cultured in the presence of the extracellular protein laminin exhibit a robust response to IGF-I and -II proteins via enhanced cell migration and angiogenic outgrowth. Furthermore, IGF vectors greatly enhanced neovascularization in an in vivo Matrigel model. Transduction of cardiomyocytes with the IGF adenoviral vectors resulted in a dose- and time-dependent increase in the expression of IGF-I or IGF-II protein. This correlated with abrogation of apoptosis induced by ischemia-reoxygenation, ceramide, or heat shock with optimal inhibition of approximately 80%. We conclude that gene transfer of IGF-I and IGF-II is a plausible strategy for the local delivery of IGFs to treat ischemic heart disease and heart failure by stimulating angiogenesis and protecting cardiomyocytes from cell death.


Subject(s)
Apoptosis , Genetic Vectors , Insulin-Like Growth Factor II/genetics , Insulin-Like Growth Factor I/genetics , Myocardium/cytology , Neovascularization, Physiologic , Adenoviridae/genetics , Animals , Aorta , Cell Adhesion , Chemotaxis , Endothelium, Vascular/cytology , Fibroblast Growth Factor 2 , Gene Expression , Genetic Therapy , Heart/embryology , Heart Diseases/therapy , Humans , Laminin/metabolism , Myocardial Ischemia/therapy , Rats , Rats, Sprague-Dawley , Recombinant Proteins , Reverse Transcriptase Polymerase Chain Reaction , Umbilical Veins
4.
Biochemistry ; 40(31): 9379-86, 2001 Aug 07.
Article in English | MEDLINE | ID: mdl-11478907

ABSTRACT

Human melanin-concentrating hormone (hMCH) is a potent but nonselective agonist at human melanin-concentrating hormone receptors 1 and 2 (hMCH-1R and hMCH-2R, respectively). To determine the structural features of this neuropeptide which are necessary for efficient binding to and activation of the receptors, Ala-substituted, open-chain, and truncated analogues were synthesized and tested in the binding assays in CHO cells expressing hMCH-1R and hMCH-2R, and in functional assays measuring the level of intracellular calcium mobilization in human HEK-293 cells expressing these receptors. A compound consisting merely of the cyclic core of hMCH with the Arg attached to the N-terminus of the disulfide ring was found to activate both hMCH-1R and hMCH-2R about as effectively as full-length hMCH. Thus, the sequence Arg-cyclo(S-S)(Cys-Met-Leu-Gly-Arg-Val-Tyr-Arg-Pro-Cys) appears to constitute the "active core" that is necessary for agonist potency at hMCH-1R and hMCH-2R. A potent and approximately 4-fold more selective agonist at hMCH-1R than at hMCH-2R is also reported.


Subject(s)
Hypothalamic Hormones/chemistry , Hypothalamic Hormones/physiology , Melanins/chemistry , Melanins/physiology , Peptide Fragments/physiology , Pituitary Hormones/chemistry , Pituitary Hormones/physiology , Receptors, Pituitary Hormone/metabolism , Alanine/metabolism , Amino Acid Sequence , Amino Acid Substitution , Cell Line , Cysteine/metabolism , Disulfides/chemistry , Disulfides/metabolism , Humans , Hypothalamic Hormones/metabolism , Isomerism , Melanins/metabolism , Molecular Sequence Data , Peptide Fragments/chemical synthesis , Peptides, Cyclic/chemical synthesis , Peptides, Cyclic/physiology , Pituitary Hormones/metabolism , Protein Conformation , Receptors, G-Protein-Coupled , Receptors, Pituitary Hormone/agonists
5.
Bioorg Med Chem Lett ; 11(14): 1955-7, 2001 Jul 23.
Article in English | MEDLINE | ID: mdl-11459669

ABSTRACT

A series of small molecules derived from MK-0677, a potent synthetic GHS, mimicking the N-terminal Gly-Ser-O-(n-octanoyl)-L-Ser-Phe segment of ghrelin was synthesized and tested in a binding and in a functional assay measuring intracellular calcium elevation in HEK-293 cells expressing hGHSR1a. Replacement of Phe in this tetrapeptide with a spiro(indoline-3,4'-piperidine) group, Gly-Ser with 2-aminoisobutyric acid, and O-(n-octanoyl)-L-Ser with O-benzyl-D-Ser provided synthetic GHS agonists with similar functional potency as ghrelin.


Subject(s)
Calcium/metabolism , Indoles/metabolism , Peptide Hormones , Peptides/metabolism , Piperidines/metabolism , Receptors, Cell Surface/metabolism , Receptors, G-Protein-Coupled , Spiro Compounds/metabolism , Binding Sites/physiology , Cells, Cultured , Ghrelin , Humans , Indoles/chemistry , Inhibitory Concentration 50 , Luminescence , Molecular Mimicry , Peptides/chemistry , Piperidines/chemical synthesis , Protein Binding/physiology , Receptors, Ghrelin , Spiro Compounds/chemistry
6.
Proc Natl Acad Sci U S A ; 98(13): 7564-9, 2001 Jun 19.
Article in English | MEDLINE | ID: mdl-11404457

ABSTRACT

Melanin-concentrating hormone (MCH) is a 19-aa cyclic neuropeptide originally isolated from chum salmon pituitaries. Besides its effects on the aggregation of melanophores in fish several lines of evidence suggest that in mammals MCH functions as a regulator of energy homeostasis. Recently, several groups reported the identification of an orphan G protein-coupled receptor as a receptor for MCH (MCH-1R). We hereby report the identification of a second human MCH receptor termed MCH-2R, which shares about 38% amino acid identity with MCH-1R. MCH-2R displayed high-affinity MCH binding, resulting in inositol phosphate turnover and release of intracellular calcium in mammalian cells. In contrast to MCH-1R, MCH-2R signaling is not sensitive to pertussis toxin and MCH-2R cannot reduce forskolin-stimulated cAMP production, suggesting an exclusive G(alpha)q coupling of the MCH-2R in cell-based systems. Northern blot and in situ hybridization analysis of human and monkey tissue shows that expression of MCH-2R mRNA is restricted to several regions of the brain, including the arcuate nucleus and the ventral medial hypothalamus, areas implicated in regulation of body weight. In addition, the human MCH-2R gene was mapped to the long arm of chromosome 6 at band 6q16.2-16.3, a region reported to be associated with cytogenetic abnormalities of obese patients. The characterization of a second mammalian G protein-coupled receptor for MCH potentially indicates that the control of energy homeostasis in mammals by the MCH neuropeptide system may be more complex than initially anticipated.


Subject(s)
Brain/metabolism , Chromosomes, Human, Pair 22 , Receptors, Pituitary Hormone/genetics , Receptors, Pituitary Hormone/metabolism , Receptors, Pituitary Hormone/physiology , Amino Acid Sequence , Animals , Base Sequence , CHO Cells , COS Cells , Cell Membrane/physiology , Chlorocebus aethiops , Chromosome Mapping , Cricetinae , Female , Humans , In Situ Hybridization , Male , Molecular Sequence Data , Oncorhynchus keta , Organ Specificity , Pituitary Gland/chemistry , Pituitary Gland/physiology , Radioligand Assay , Receptors, G-Protein-Coupled , Receptors, Pituitary Hormone/chemistry , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Transfection
7.
Mol Pharmacol ; 59(4): 867-74, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11259632

ABSTRACT

In the present study, rat cardiac myocytes were used as an in vitro ischemia/reperfusion injury model to delineate the role of c-Jun N-terminal kinase (JNK) 1 and JNK2 isoforms in ischemia/reoxygenation-induced apoptosis using an antisense approach. Exposure of rat cardiac myocytes to ischemia did not induce apoptosis as detected by staining with either acridine orange/ethidium bromide or annexin-V-fluorescein/propidium iodide. In contrast, a time-dependent increase in the number of apoptotic cells was noted after reoxygenation of ischemic myocytes, whereas the level of necrotic cells remained unaltered. Reoxygenation, but not ischemia alone, also caused a time-dependent increase in JNK activation that preceded apoptosis induction. Treatment of cardiac myocytes with antisense (AS) oligonucleotides that specifically targeted either JNK1 or JNK2 significantly reduced both mRNA and protein expression of the target isoform but had no effect on the expression of the alternate isoform. Pretreatment of cardiac myocytes with JNK1 AS, but not JNK2 AS, resulted in almost complete attenuation of reoxygenation-induced apoptosis. Furthermore, control oligonucleotides for JNK1 AS or JNK2 AS had no effect on JNK mRNA or protein expression or reoxygenation-induced apoptosis, indicating a sequence-specific mode of action. Additional studies revealed that apoptosis induced by other JNK-activating stimuli, including ceramide, heat shock, and UV irradiation, was partly suppressed after treatment with JNK1 AS but not JNK2 AS. These findings demonstrate that the JNK1 isoform plays a preferential role in apoptosis induced by ischemia/reoxygenation as well as diverse JNK-activating cellular stresses.


Subject(s)
Apoptosis/drug effects , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Myocardial Ischemia/metabolism , Myocardium/metabolism , Animals , Apoptosis/radiation effects , Cell Hypoxia/drug effects , Cells, Cultured , Enzyme Induction/physiology , Heat-Shock Response/drug effects , Isoenzymes/antagonists & inhibitors , Mitogen-Activated Protein Kinase 8 , Mitogen-Activated Protein Kinase 9 , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Myocardium/cytology , Oligonucleotides, Antisense/pharmacology , Oxygen/pharmacology , Phosphorylation/drug effects , Phosphorylation/radiation effects , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Reperfusion Injury/metabolism , Ultraviolet Rays
8.
J Med Chem ; 43(23): 4370-6, 2000 Nov 16.
Article in English | MEDLINE | ID: mdl-11087562

ABSTRACT

The recently discovered growth hormone secretagogue, ghrelin, is a potent agonist at the human growth hormone secretagogue receptor 1a (hGHSR1a). To elucidate structural features of this peptide necessary for efficient binding to and activation of the receptor, several analogues of ghrelin with various aliphatic or aromatic groups in the side chain of residue 3, and several short peptides derived from ghrelin, were prepared and tested in a binding assay and in an assay measuring intracellular calcium elevation in HEK-293 cells expressing hGHSR1a. Bulky hydrophobic groups in the side chain of residue 3 turned out to be essential for maximum agonist activity. Also, short peptides encompassing the first 4 or 5 residues of ghrelin were found to functionally activate hGHSR1a about as efficiently as the full-length ghrelin. Thus the entire sequence of ghrelin is not necessary for activity: the Gly-Ser-Ser(n-octanoyl)-Phe segment appears to constitute the "active core" required for agonist potency at hGHSR1a.


Subject(s)
Peptide Hormones , Peptides/chemistry , Receptors, Cell Surface/agonists , Receptors, G-Protein-Coupled , Amino Acid Sequence , Binding Sites , Calcium/metabolism , Cell Line , Ghrelin , Humans , Luminescent Measurements , Magnetic Resonance Spectroscopy , Molecular Sequence Data , Peptide Fragments/chemical synthesis , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Peptides/metabolism , Peptides/pharmacology , Receptors, Cell Surface/metabolism , Receptors, Ghrelin , Structure-Activity Relationship
9.
Nature ; 406(6791): 70-4, 2000 Jul 06.
Article in English | MEDLINE | ID: mdl-10894543

ABSTRACT

Neuromedin U (NMU) is a neuropeptide with potent activity on smooth muscle which was isolated first from porcine spinal cord and later from other species. It is widely distributed in the gut and central nervous system. Peripheral activities of NMU include stimulation of smooth muscle, increase of blood pressure, alteration of ion transport in the gut, control of local blood flow and regulation of adrenocortical function. An NMU receptor has not been molecularly identified. Here we show that the previously described orphan G-protein-coupled receptor FM-3 (ref. 15) and a newly discovered one (FM-4) are cognate receptors for NMU. FM-3, designated NMU1R, is abundantly expressed in peripheral tissues whereas FM-4, designated NMU2R, is expressed in specific regions of the brain. NMU is expressed in the ventromedial hypothalamus in the rat brain, and its level is significantly reduced following fasting. Intracerebroventricular administration of NMU markedly suppresses food intake in rats. These findings provide a molecular basis for the biochemical activities of NMU and may indicate that NMU is involved in the central control of feeding.


Subject(s)
Feeding Behavior/physiology , Membrane Proteins , Neuropeptides/metabolism , Receptors, Cell Surface/physiology , Receptors, Neurotransmitter/physiology , Amino Acid Sequence , Animals , Brain/metabolism , Cell Line , Cloning, Molecular , Fasting , Humans , Ligands , Mice , Molecular Sequence Data , Obesity/metabolism , RNA, Messenger/metabolism , Radioligand Assay , Rats , Receptors, Cell Surface/analysis , Receptors, Neurotransmitter/analysis , Sequence Alignment , Tissue Distribution
10.
Mol Endocrinol ; 14(1): 160-9, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10628755

ABSTRACT

Synthetic ligands have been identified that reset and amplify the cycle of pulsatile GH secretion by interacting with the orphan GH-secretagogue receptor (GHS-R). The GHS-R is rhodopsin like, but does not obviously belong to any of the established G protein-coupled receptor (GPCR) subfamilies. We recently characterized the closely related orphan family member, GPR38, as the motilin receptor. A common property of both receptors is that they amplify and sustain pulsatile biological responses in the continued presence of their respective ligands. To efficiently identify additional members of this new GPCR family, we explored a vertebrate species having a compact genome, that was evolutionary distant from human, but where functionally important genes were likely to be conserved. Accordingly, three distinct full-length clones, encoding proteins of significant identity to the human GHS-R, were isolated from the Pufferfish (Spheroides nephelus). Southern analyses showed that the three cloned Pufferfish genes are highly conserved across species. The gene with closest identity (58%) was activated by three synthetic ligands that were chosen for their very high selectivity on the GHS-R as illustrated by their specificity in activating the wild-type human GHS-R but not the E124Q mutant. These results indicate that the ligand activation domain of the GHS-R has been evolutionary conserved from Pufferfish to human (400 million years), supporting the notion that the GHS-R and its natural ligand play a fundamentally important role in biology. Furthermore, they illustrate the power of exploiting the compact Pufferfish genome for simplifying the isolation of endocrinologically important receptor families.


Subject(s)
Fishes/genetics , Receptors, Cell Surface/chemistry , Receptors, G-Protein-Coupled , Amino Acid Sequence , Animals , Blotting, Southern , Cell Line , Cloning, Molecular , Conserved Sequence , Genomic Library , Humans , Ligands , Models, Genetic , Molecular Sequence Data , Protein Structure, Tertiary , Receptors, Cell Surface/genetics , Receptors, Ghrelin , Sequence Alignment , Sequence Homology, Amino Acid , Transfection
11.
Horm Res ; 51 Suppl 3: 1-8, 1999.
Article in English | MEDLINE | ID: mdl-10592437

ABSTRACT

A series of structurally diverse growth hormone (GH) releasing substances have been synthesized that are distinct from the naturally occurring GH releasing hormone (GHRH). These synthetic molecules range from the family of GH releasing peptides and mimetics such as MK-0677. The physiological importance of these molecules and their receptor is exemplified by studies in the elderly. For example, when MK-0677 was administered chronically to 70- to 90-year-old subjects, once daily, the age-related reduced amplitude of GH pulses was reversed to that of the physiological profile typical of young adults. In 1996, the synthesis of (35)S-MK-0677 was reported and used as a ligand to characterize a common receptor (GH secretagogue receptor [GHS-R]) for the GH releasing substances. The GHS-R is distinct from the GHRH receptor. Subsequently, the GHS-R gene was cloned and shown to encode a unique G-protein coupled receptor with a deduced protein sequence that was 96% identical in human and rat. Because of the physiological importance of the GHS-R, a search for family members (FMs) was initiated and its molecular evolution investigated. Three FMs GPR38, GPR39 and FM3 were isolated from human genomic libraries. To accelerate the identification of other FMs, a vertebrate organism with a compact genome distant in evolutionary terms from humans was exploited. The pufferfish (Spheroides nephelus) genome provides an ideal model for the discovery of human genes. Three distinct full-length clones encoding proteins of significant sequence identity to the human GHS-R were cloned from the pufferfish. Remarkably, the pufferfish gene with highest sequence homology to the human receptor was activated by the hexapeptide and non-peptide ligands. These intriguing results show that the structure and function of the ligand binding pocket of the human GHS-R has been highly conserved in evolution ( approximately 400 million years) and strongly suggests that an endogenous natural ligand has been conserved. This new information is consistent with a natural ligand for the GHS-R playing a fundamentally important and conserved role in physiology.


Subject(s)
Growth Hormone-Releasing Hormone , Receptors, Cell Surface/physiology , Receptors, G-Protein-Coupled , Aged , Aged, 80 and over , Amino Acid Sequence , Animals , Binding Sites , Human Growth Hormone/metabolism , Humans , Indoles/pharmacology , Molecular Sequence Data , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/metabolism , Receptors, Ghrelin , Receptors, Neuropeptide , Receptors, Pituitary Hormone-Regulating Hormone , Spiro Compounds/pharmacology
12.
FEBS Lett ; 451(2): 137-41, 1999 May 21.
Article in English | MEDLINE | ID: mdl-10371153

ABSTRACT

The primary hormonal regulator of pigmentation is melanocyte stimulating hormone derived from proopiomelanocortin by proteolytic processing. The melanocortin-1 receptor serves a key role in the regulation of pigmentation. We describe the identification of the first intron within a melanocortin receptor. A new melanocortin-1 receptor isoform, generated by alternative mRNA splicing, encodes an additional 65 amino acids at the predicted intracellular, C-terminal tail of the melanocortin-1 receptor. When expressed in heterologous cells, the new spliced form of the melanocortin-1 receptor (melanocortin-1 receptor B) appears pharmacologically similar to the non-spliced melanocortin-1 receptor. Melanocortin-1 receptor B is expressed in testis, fetal heart and melanomas.


Subject(s)
Alternative Splicing , Receptors, Corticotropin/genetics , Amino Acid Sequence , Animals , Base Sequence , CHO Cells , Cloning, Molecular , Cricetinae , Expressed Sequence Tags , Humans , Inhibitory Concentration 50 , Models, Genetic , Molecular Sequence Data , Polymorphism, Genetic , Protein Binding , Receptors, Corticotropin/metabolism , Receptors, Melanocortin
13.
Science ; 284(5423): 2184-8, 1999 Jun 25.
Article in English | MEDLINE | ID: mdl-10381885

ABSTRACT

Motilin is a 22-amino acid peptide hormone expressed throughout the gastrointestinal (GI) tract of humans and other species. It affects gastric motility by stimulating interdigestive antrum and duodenal contractions. A heterotrimeric guanosine triphosphate-binding protein (G protein)-coupled receptor for motilin was isolated from human stomach, and its amino acid sequence was found to be 52 percent identical to the human receptor for growth hormone secretagogues. The macrolide antibiotic erythromycin also interacted with the cloned motilin receptor, providing a molecular basis for its effects on the human GI tract. The motilin receptor is expressed in enteric neurons of the human duodenum and colon. Development of motilin receptor agonists and antagonists may be useful in the treatment of multiple disorders of GI motility.


Subject(s)
Colon/metabolism , Gastric Mucosa/metabolism , Intestine, Small/metabolism , Motilin/metabolism , Receptors, Gastrointestinal Hormone/chemistry , Receptors, Gastrointestinal Hormone/genetics , Receptors, Neuropeptide/chemistry , Receptors, Neuropeptide/genetics , Alternative Splicing , Amino Acid Sequence , Base Sequence , Binding Sites , Calcium/metabolism , Cell Line , Chromosome Mapping , Chromosomes, Human, Pair 13 , Cloning, Molecular , Erythromycin/metabolism , GTP-Binding Proteins/metabolism , Humans , In Situ Hybridization , Ligands , Molecular Sequence Data , Motilin/analogs & derivatives , Receptors, Gastrointestinal Hormone/metabolism , Receptors, Neuropeptide/metabolism , Thyroid Gland/metabolism , Transfection
14.
J Neurochem ; 71(6): 2239-51, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9832121

ABSTRACT

Galanin is a 29- or 30-amino acid peptide with wide-ranging effects on hormone release, feeding behavior, smooth muscle contractility, and somatosensory neuronal function. Three distinct galanin receptor (GALR) subtypes, designated GALR1, 2, and 3, have been cloned from the rat. We report here the cloning of the human GALR2 and GALR3 genes, an initial characterization of their pharmacology with respect to radioligand binding and signal transduction pathways, and a profile of their expression in brain and peripheral tissues. Human GALR2 and GALR3 show, respectively, 92 and 89% amino acid sequence identity with their rat homologues. Radioligand binding studies with 125I-galanin show that recombinant human GALR2 binds with high affinity to human galanin (K(D) = 0.3 nM). Human GALR3 binds galanin with less affinity (IC50 of 12 nM for porcine galanin and 75 nM for human galanin). Human GALR2 was shown to couple to phospholipase C and elevation of intracellular calcium levels as assessed by aequorin luminescence in HEK-293 cells and by Xenopus melanophore pigment aggregation and dispersion assays, in contrast to human GALR1 and human GALR3, which signal predominantly through inhibition of adenylate cyclase. GALR2 mRNA shows a wide distribution in the brain (mammillary nuclei, dentate gyrus, cingulate gyrus, and posterior hypothalamic, supraoptic, and arcuate nuclei), and restricted peripheral tissue distribution with highest mRNA levels detected in human small intestine. In comparison, whereas GALR3 mRNA was expressed in many areas of the rat brain, there was abundant expression in the primary olfactory cortex, olfactory tubercle, the islands of Calleja, the hippocampal CA regions of Ammon's horn, and the dentate gyrus. GALR3 mRNA was highly expressed in human testis and was detectable in adrenal gland and pancreas. The genes for human GALR2 and 3 were localized to chromosomes 17q25 and 22q12.2-13.1, respectively.


Subject(s)
Cloning, Molecular , Receptors, Neuropeptide/genetics , Receptors, Neuropeptide/metabolism , Amino Acid Sequence , Animals , Blotting, Northern , Brain/metabolism , Cell Line , Humans , Isomerism , Ligands , Mice , Molecular Sequence Data , Nucleic Acid Hybridization , Rats , Receptors, Galanin , Receptors, Neuropeptide/physiology , Ribonucleases , Signal Transduction/physiology , Swine , Xenopus laevis
15.
Genomics ; 52(2): 223-9, 1998 Sep 01.
Article in English | MEDLINE | ID: mdl-9782091

ABSTRACT

Growth hormone secretagogues (GHS) are a group of synthetic peptide and nonpeptide molecules that potently stimulate the release of GH from the anterior pituitary gland through the activation of a novel G-protein-coupled receptor (GPC-R), the GHS-R. In our search for GHS-R family members, we recently described the cloning of two related GPC-Rs, GPR38 and 39. In the present report, we detail the isolation of a new GPC-R (FM-3) from human and mouse with moderate sequence identity to both the GHS-R and neurotensin-R. FM-3 is expressed in a diverse set of tissues.


Subject(s)
GTP-Binding Proteins/metabolism , Membrane Proteins , Receptors, Cell Surface/genetics , Receptors, G-Protein-Coupled , Receptors, Neurotensin/genetics , Receptors, Neurotransmitter , T-Lymphocytes/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cell Line , Cloning, Molecular , DNA/chemistry , DNA/genetics , DNA, Complementary/chemistry , DNA, Complementary/genetics , Gene Expression , Humans , Mice , Molecular Sequence Data , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Cell Surface/metabolism , Receptors, Ghrelin , Sequence Analysis, DNA , Sequence Homology, Amino Acid , T-Lymphocytes/chemistry , Tissue Distribution , Transfection
16.
Mol Endocrinol ; 12(1): 137-45, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9440817

ABSTRACT

Antibodies raised against an intracellular and extracellular domain of the GH secretagogue receptor (GHS-R) confirmed that its topological orientation in the lipid bilayer is as predicted for G protein-coupled receptors with seven transmembrane domains. A strategy for mapping the agonist-binding site of the human GHS-R was conceived based on our understanding of ligand binding in biogenic amine and peptide hormone G protein-coupled receptors. Using site-directed mutagenesis and molecular modeling, we classified GHS peptide and nonpeptide agonist binding in the context of its receptor environment. All peptide and nonpeptide ligand classes shared a common binding domain in transmembrane (TM) region 3 of the GHS-R. This finding was based on TM-3 mutation E124Q, which eliminated the counter-ion to the shared basic N+ group of all GHSs and resulted in a nonfunctional receptor. Restoration of function for the E124Q mutant was achieved by a complementary change in the MK-0677 ligand through modification of its amine side-chain to the corresponding alcohol. Contacts in other TM domains [TM-2 (D99N), TM-5 (M213K, S117A), TM-6 (H280F), and extracellular loop 1 (C116A)] of the receptor revealed specificity for the different peptide, benzolactam, and spiroindolane GHSs. GHS-R agonism, therefore, does not require identical disposition of all agonist classes at the ligand-binding site. Our results support the hypothesis that the ligand-binding pocket in the GHS-R is spatially disposed similarly to the well characterized catechol-binding site in the beta2-adrenergic receptor.


Subject(s)
GTP-Binding Proteins/chemistry , GTP-Binding Proteins/metabolism , Human Growth Hormone/metabolism , Peptides/metabolism , Peptides/physiology , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/metabolism , Receptors, G-Protein-Coupled , Amino Acid Sequence , Animals , GTP-Binding Proteins/genetics , GTP-Binding Proteins/physiology , Human Growth Hormone/chemistry , Human Growth Hormone/genetics , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Peptides/chemistry , Peptides/genetics , Rats , Receptors, Cell Surface/genetics , Receptors, Ghrelin , Swine
17.
Can J Physiol Pharmacol ; 75(6): 735-40, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9276157

ABSTRACT

We developed a functional assay to evaluate the activity of kinin peptides at the human and mouse bradykinin (BK) B1 receptors. The photoprotein aequorin expressed in 293-AEQ17 cells was used to measure calcium mobilization due to activation of the human or mouse B1 receptors by kinin peptides. The B1 agonists des-Arg9-BK and des-Arg10-kallidin activated the human receptor (EC50 = 112 and 5 nM, respectively), whereas the B1 peptide antagonists des-Arg9,Leu8-BK and des-Arg10,Leu9-kallidin showed no activation. At the murine receptor, the agonists des-Arg9-BK and des-Arg10-kallidin activated the receptor with EC50 values of 39 and 23 nM, respectively. In contrast, the two peptide antagonists showed significant agonism at the murine receptor. Thirty-nine and 44% agonism of the mouse receptor was observed with des-Arg9,Leu8-BK (EC50 = 56 nM) and des-Arg10,Leu9-kallidin (EC50 = 177 nM). Two recently described kinin analogues, [Lys-Lys0,Hyp3,Igl5,D-Igl7,Oic8,des-Arg9]B K and [D-Arg0,Hyp3,Igl5,D-Igl7, Oic8,des-Arg9]BK (B9858 and des-Arg9-B9430), failed to agonize the mouse receptor. These peptides were potent antagonists of des-Arg10-kallidin- and des-Arg9-BK-induced bioluminescence at the cloned human and mouse B1 receptors.


Subject(s)
Bradykinin Receptor Antagonists , Bradykinin/analogs & derivatives , Peptides/pharmacology , Receptors, Bradykinin/agonists , Animals , Bradykinin/pharmacology , COS Cells/metabolism , Humans , Kinetics , Mice , Peptides/metabolism , Receptor, Bradykinin B1 , Receptor, Bradykinin B2 , Receptors, Bradykinin/metabolism , Species Specificity
18.
Mol Endocrinol ; 11(4): 415-23, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9092793

ABSTRACT

GH release is thought to occur under the reciprocal regulation of two hypothalamic peptides, GH releasing hormone (GHRH) and somatostatin, via their engagement with specific cell surface receptors on the anterior pituitary somatotroph. In addition, GH-releasing peptides, such as GHRP-6 and the nonpeptide mimetics, L-692,429 and MK-0677, stimulate GH release through their activation of a distinct receptor, the GH secretagogue receptor (GHS-R). The recent cloning of the GHS-R from human and swine pituitary gland identifies yet a third G protein-coupled receptor (GPC-R) involved in the control of GH release and further supports the existence of an undiscovered hormone that may activate this receptor. Using the human GHS-R as a probe, we report the isolation of a rat pituitary GHS-R cDNA derived from an unspliced, precursor mRNA. The rat cDNA encodes a protein of 364 amino acids containing seven transmembrane domains (7-TM) with >90% sequence identity to both the human and swine GHS-Rs. A single intron of approximately 2 kb divides the open reading frame into two exons encoding TM 1-5 and TM 6-7, thus placing the GHS-R into the intron-containing class of GPC-Rs. The intron maps to the site of sequence divergence between the human and swine type 1a and 1b GHS-R mRNAs. In addition, determination of the nucleotide sequence for the human GHS-R gene confirmed the position of an intron in the human GHS-R gene at this position. A full-length contiguous cDNA from rat hypothalamus was isolated and shown to be identical in its nucleotide and deduced amino acid sequence to the rat pituitary GHS-R. The cloned rat GHS-R binds [35S]MK-0677 with high affinity [dissociation constant (K(D)) = 0.7 nM] and is functionally active when expressed in HEK-293 cells. Expression of the rat GHS-R was observed specifically in the pituitary and hypothalamus when compared with control tissues.


Subject(s)
Hypothalamus/metabolism , Pituitary Gland/metabolism , Receptors, Cell Surface/genetics , Receptors, G-Protein-Coupled , Receptors, Somatotropin/genetics , Amino Acid Sequence , Animals , Base Sequence , Cell Line , Cloning, Molecular , Conserved Sequence , DNA, Complementary , Humans , In Situ Hybridization , Indoles/metabolism , Introns , Kinetics , Molecular Sequence Data , Open Reading Frames , RNA Precursors , Rats , Receptors, Ghrelin , Receptors, Somatotropin/biosynthesis , Sequence Alignment , Spiro Compounds/metabolism , Swine , Transfection
19.
Mol Pharmacol ; 51(3): 383-9, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9058592

ABSTRACT

Raf kinases, cytoplasmic serine/threonine protein kinases, have been proposed as important participants in mitogen-induced signal transduction. However, the precise role that Raf kinase isozymes play in cellular responses such as proliferation has not been resolved. The present study investigates the ability of antisense phosphorothioate oligodeoxynucleotides (ODNs), targeted against rat C-Raf and A-Raf kinases, to reduce gene expression and proliferation of cultured rat A10 smooth muscle cells (SMCs). Exposure of A10 cells to ISIS 11061, an active C-Raf antisense ODN, resulted in a potent, dose-dependent inhibition (IC50 = 55 nM) of C-Raf mRNA and protein expression. This inhibition was completely dependent on ODN sequence because the incorporation of increasing numbers of mismatches (up to six) into the sequence resulted in sequential loss of potency. Similarly, a dose-dependent reduction (IC50 = 125 nM) in A-Raf gene expression was observed after treatment of cells with the active A-Raf ODN, ISIS 9069, whereas two scrambled controls were without effect. These results demonstrate that ISIS 11061 and ISIS 9069 reduced gene expression in a sequence-specific and isozyme-specific manner. Moreover, administration of ISIS 11061 and ISIS 9069 to rat SMCs resulted in a significant and potent diminution of serum-induced proliferation with corresponding IC50 values of 216 and 273 nM, respectively. Taken together, these results indicate that A-Raf and C-Raf kinases play an important role in regulating vascular SMC proliferation and that antisense-mediated inhibition of Raf kinase activity may serve as a therapeutic modality in the treatment of vascular proliferative disorders.


Subject(s)
Gene Expression Regulation , Muscle, Smooth, Vascular/enzymology , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Animals , Cell Division , Cell Line , Isoenzymes/metabolism , Molecular Sequence Data , Oligonucleotides, Antisense/pharmacology , Proto-Oncogene Proteins c-raf , RNA, Messenger/metabolism , Rats
20.
Genomics ; 46(3): 426-34, 1997 Dec 15.
Article in English | MEDLINE | ID: mdl-9441746

ABSTRACT

The recent cloning of a growth hormone secretagogue receptor (GHS-R) from human pituitary gland and brain identified a third G protein-coupled receptor (GPC-R) involved in the control of growth hormone release. The nucleotide sequence of the GHS-R is most closely related to the neurotensin receptor-1 (NT-R1) (35% overall protein identity). Two human GPC-Rs related to both the type 1a GHS-R and NT-Rs were cloned and characterized. Hybridization at low posthybridizational stringency with restriction enzyme-digested human genomic DNA resulted in the identification of a genomic clone encoding a first GHS-R/NT-R family member (GPR38). A cDNA clone was identified encoding a second GHS-R-related gene (GPR39). GPR38 and GPR39 share significant amino acid sequence identity with the GHS-R and NT-Rs 1 and 2. An acidic residue (E124) in TM-3, essential for the binding and activation of the GHS-R by structurally dissimilar GHSs, was conserved in GPR38 and GPR39. GPR38 is encoded by a single gene expressed in thyroid gland, stomach, and bone marrow. GPR39 is encoded by a highly conserved single-copy gene, expressed in brain and other peripheral tissues. Fluorescence in situ hybridization localized the genes for GPR38 and GPR39 to separate chromosomes, distinct from the gene encoding the GHS-R and NT-R type 1. The ligand-binding and functional properties of GPR38 and GPR39 remain to be determined.


Subject(s)
Chromosomes, Human, Pair 13 , Chromosomes, Human, Pair 2 , GTP-Binding Proteins/metabolism , Receptors, Cell Surface/genetics , Receptors, G-Protein-Coupled , Receptors, Neurotensin/genetics , Amino Acid Sequence , Animals , Chromosome Mapping , Cloning, Molecular , Conserved Sequence , Evolution, Molecular , Gene Expression , Humans , Molecular Sequence Data , Receptors, Ghrelin , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...