Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 114(9): 2389-2394, 2017 02 28.
Article in English | MEDLINE | ID: mdl-28193887

ABSTRACT

Parkinson disease (PD) is a neurodegenerative disorder pathologically characterized by nigrostriatal dopamine neuron loss and the postmortem presence of Lewy bodies, depositions of insoluble α-synuclein, and other proteins that likely contribute to cellular toxicity and death during the disease. Genetic and biochemical studies have implicated impaired lysosomal and mitochondrial function in the pathogenesis of PD. Transmembrane protein 175 (TMEM175), the lysosomal K+ channel, is centered under a major genome-wide association studies peak for PD, making it a potential candidate risk factor for the disease. To address the possibility that variation in TMEM175 could play a role in PD pathogenesis, TMEM175 function was investigated in a neuronal model system. Studies confirmed that TMEM175 deficiency results in unstable lysosomal pH, which led to decreased lysosomal catalytic activity, decreased glucocerebrosidase activity, impaired autophagosome clearance by the lysosome, and decreased mitochondrial respiration. Moreover, TMEM175 deficiency in rat primary neurons resulted in increased susceptibility to exogenous α-synuclein fibrils. Following α-synuclein fibril treatment, neurons deficient in TMEM175 were found to have increased phosphorylated and detergent-insoluble α-synuclein deposits. Taken together, data from these studies suggest that TMEM175 plays a direct and critical role in lysosomal and mitochondrial function and PD pathogenesis and highlight this ion channel as a potential therapeutic target for treating PD.


Subject(s)
Autophagosomes/metabolism , Dopaminergic Neurons/metabolism , Lysosomes/metabolism , Mitochondria/metabolism , Potassium Channels/genetics , alpha-Synuclein/chemistry , Animals , Autophagosomes/drug effects , Autophagosomes/pathology , Autophagy/drug effects , Cell Line, Tumor , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/pathology , Gene Expression Regulation , Glucosylceramidase/genetics , Glucosylceramidase/metabolism , Humans , Hydrogen-Ion Concentration , Lysosomes/drug effects , Lysosomes/pathology , Mitochondria/drug effects , Mitochondria/pathology , Models, Biological , Parkinson Disease/genetics , Parkinson Disease/metabolism , Parkinson Disease/pathology , Potassium Channels/deficiency , Primary Cell Culture , Protein Aggregates/drug effects , Rats , alpha-Synuclein/pharmacology
2.
Mol Ther Nucleic Acids ; 5(8): e342, 2016 Aug 02.
Article in English | MEDLINE | ID: mdl-27483025

ABSTRACT

Short interfering RNAs (siRNAs) are a valuable tool for gene silencing with applications in both target validation and therapeutics. Many advances have recently been made to improve potency and specificity, and reduce toxicity and immunostimulation. However, siRNA delivery to a variety of tissues remains an obstacle for this technology. To date, siRNA delivery to muscle has only been achieved by local administration or by methods with limited potential use in the clinic. We report systemic delivery of a highly chemically modified cholesterol-conjugated siRNA targeting muscle-specific gene myostatin (Mstn) to a full range of muscles in mice. Following a single intravenous injection, we observe 85-95% knockdown of Mstn mRNA in skeletal muscle and >65% reduction in circulating Mstn protein sustained for >21 days. This level of Mstn knockdown is also accompanied by a functional effect on skeletal muscle, with animals showing an increase in muscle mass, size, and strength. The cholesterol-conjugated siRNA platform described here could have major implications for treatment of a variety of muscle disorders, including muscular atrophic diseases, muscular dystrophy, and type II diabetes.

3.
Mol Ther Nucleic Acids ; 4: e224, 2015 Jan 27.
Article in English | MEDLINE | ID: mdl-25625614

ABSTRACT

The present study aimed at establishing feasibility of delivering short interfering RNA (siRNA) to target the coagulation cascade in rat and rabbit, two commonly used species for studying thrombosis and hemostasis. siRNAs that produced over 90% mRNA knockdown of rat plasma prekallikrein and rabbit Factor X (FX) were identified from in vitro screens. An ionizable amino lipid based lipid nanoparticle (LNP) formulation for siRNA in vivo delivery was characterized as tolerable and exerting no appreciable effect on coagulability at day 7 postdosing in both species. Both prekallikrein siRNA-LNP and FX siRNA-LNP resulted in dose-dependent and selective knockdown of target gene mRNA in the liver with maximum reduction of over 90% on day 7 following a single dose of siRNA-LNP. Knockdown of plasma prekallikrein was associated with modest clot weight reduction in the rat arteriovenous shunt thrombosis model and no increase in the cuticle bleeding time. Knockdown of FX in the rabbit was accompanied with prolongation in ex vivo clotting times. Results fit the expectations with both targets and demonstrate for the first time, the feasibility of targeting coagulation factors in rat, and, more broadly, targeting a gene of interest in rabbit, via systemic delivery of ionizable LNP formulated siRNA.

4.
Bioorg Med Chem Lett ; 18(9): 2860-4, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18420409

ABSTRACT

A series of 2-aminomethyl piperidines has been discovered as novel urotensin-II receptor antagonists. The synthesis, initial structure-activity relationships, and optimization of the initial hit that resulted in the identification of potent, cross-species active, and functional urotensin-II receptor antagonists such as 1a and 11a are described.


Subject(s)
Methylamines/pharmacology , Piperidines/pharmacology , Receptors, G-Protein-Coupled/antagonists & inhibitors , Vasoconstrictor Agents/pharmacology , Binding Sites , Humans , Methylamines/chemical synthesis , Models, Chemical , Piperidines/chemical synthesis , Stereoisomerism , Structure-Activity Relationship , Vasoconstrictor Agents/chemical synthesis
5.
Mol Cell Biochem ; 295(1-2): 113-20, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16896535

ABSTRACT

Lysophosphatidylcholine (LPC) is the major bioactive lipid component of oxidized LDL, thought to be responsible for many of the inflammatory effects of oxidized LDL described in both inflammatory and endothelial cells. Inflammation-induced transformation of vascular smooth muscle cells from a contractile phenotype to a proliferative/secretory phenotype is a hallmark of the vascular remodeling that is characteristic of atherogenesis; however, the role of LPC in this process has not been fully described. The present study tested the hypothesis that LPC is an inflammatory stimulus in coronary artery smooth muscle cells (CASMCs). In cultured human CASMCs, LPC stimulated time- and concentration-dependent release of arachidonic acid that was sensitive to phospholipase A2 and C inhibition. LPC stimulated the release of arachidonic acid metabolites leukotriene-B4 and 6-keto-prostaglandin F1alpha, within the same time course. LPC was also found to stimulate basic fibroblast growth factor release as well as stimulating the release of the cytokines GM-CSF, IL-6, and IL-8. Optimal stimulation of these signals was obtained via palmitic acid-substituted LPC species. Stimulation of arachidonic acid, inflammatory cytokines and growth factor release, implies that LPC might play a multifactorial role in the progression of atherosclerosis, by affecting inflammatory processes.


Subject(s)
Coronary Vessels/cytology , Coronary Vessels/drug effects , Inflammation/pathology , Lysophosphatidylcholines/pharmacology , Myocytes, Smooth Muscle/drug effects , 6-Ketoprostaglandin F1 alpha/biosynthesis , Arachidonic Acid/metabolism , Arachidonic Acids/pharmacology , Cells, Cultured , Cytokines/metabolism , Fibroblast Growth Factor 2/biosynthesis , Humans , Leukotriene B4/biosynthesis , Lipids/pharmacology , Myocytes, Smooth Muscle/metabolism , Phospholipases/antagonists & inhibitors , Tritium
6.
Peptides ; 27(6): 1532-7, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16290260

ABSTRACT

Urotensin-II (U-II), a ligand for the G-protein-coupled receptor UT, has been characterized as the most potent mammalian vasoconstrictor identified to date. Although circulating levels of U-II are altered in lower species (e.g., fish) upon exposure to hypo-osmotic stress, little is known about the actions of this cyclic undecapeptide within the kidney, an organ that plays a pivotal role in the control of cardiovascular homeostasis, influencing both cardiac preload (plasma volume) and after load (peripheral resistance). The present study reports the identification of specific, high affinity [125I]hU-II binding sites in Sprague-Dawley rat kidney outer medulla by autoradiography and also through membrane radioligand binding (Kd 1.9 +/- 0.9 nM and Bmax 408 +/- 47 amol mm(-2) and Kd 1.4 +/- 0.3 nM and Bmax 51.3 +/- 7.8 fmol mg(-1) protein, respectively). Differences were observed in the binding characteristics within rat strains. Compared to the Sprague-Dawley, Wistar Kyoto (WKY) and spontaneously hypertensive (SHR) rat kidney outer medulla displayed low density < 20 fmol mg(-1) protein and low affinity (> 1 microM) [125I]hU-II binding sites. Thus, the relative contribution of specific U-II binding sites to the physiological actions of U-II in the control of cardiorenal homeostasis is worthy of further investigation.


Subject(s)
Kidney Medulla/metabolism , Urotensins/chemistry , Animals , Autoradiography , Binding Sites , Binding, Competitive , Humans , Kinetics , Ligands , Protein Binding , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/metabolism , Species Specificity , Urotensins/metabolism
7.
Br J Pharmacol ; 145(5): 620-35, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15852036

ABSTRACT

1. SB-706375 potently inhibited [(125)I]hU-II binding to both mammalian recombinant and 'native' UT receptors (K(i) 4.7+/-1.5 to 20.7+/-3.6 nM at rodent, feline and primate recombinant UT receptors and K(i) 5.4+/-0.4 nM at the endogenous UT receptor in SJRH30 cells). 2. Prior exposure to SB-706375 (1 microM, 30 min) did not alter [(125)I]hU-II binding affinity or density in recombinant cells (K(D) 3.1+/-0.4 vs 5.8+/-0.9 nM and B(max) 3.1+/-1.0 vs 2.8+/-0.8 pmol mg(-1)) consistent with a reversible mode of action. 3. The novel, nonpeptidic radioligand [(3)H]SB-657510, a close analogue of SB-706375, bound to the monkey UT receptor (K(D) 2.6+/-0.4 nM, B(max) 0.86+/-0.12 pmol mg(-1)) in a manner that was inhibited by both U-II isopeptides and SB-706375 (K(i) 4.6+/-1.4 to 17.6+/-5.4 nM) consistent with the sulphonamides and native U-II ligands sharing a common UT receptor binding domain. 4. SB-706375 was a potent, competitive hU-II antagonist across species with pK(b) 7.29-8.00 in HEK293-UT receptor cells (inhibition of [Ca(2+)](i)-mobilization) and pK(b) 7.47 in rat isolated aorta (inhibition of contraction). SB-706375 also reversed tone established in the rat aorta by prior exposure to hU-II (K(app) approximately 20 nM). 5. SB-706375 was a selective U-II antagonist with >/=100-fold selectivity for the human UT receptor compared to 86 distinct receptors, ion channels, enzymes, transporters and nuclear hormones (K(i)/IC(50)>1 microM). Accordingly, the contractile responses induced in isolated aortae by KCl, phenylephrine, angiotensin II and endothelin-1 were unaltered by SB-706375 (1 microM). 6. In summary, SB-706375 is a high-affinity, surmountable, reversible and selective nonpeptide UT receptor antagonist with cross-species activity that will assist in delineating the pathophysiological actions of U-II in mammals.


Subject(s)
Pyrrolidines/pharmacology , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/genetics , Sulfonamides/pharmacology , Algorithms , Animals , Aorta, Thoracic/drug effects , Binding, Competitive/drug effects , Cats , Cell Line, Tumor , Cell Membrane/metabolism , Haplorhini , Humans , In Vitro Techniques , Mice , Muscle Contraction/drug effects , Muscle, Smooth, Vascular/drug effects , Radioligand Assay , Rats , Recombinant Proteins/metabolism , Rhabdomyosarcoma/metabolism , Species Specificity
8.
Biochem Pharmacol ; 69(7): 1069-79, 2005 Apr 01.
Article in English | MEDLINE | ID: mdl-15763543

ABSTRACT

Urotensin-II (U-II), acting through its G-protein-coupled receptor, UT, is a possible contributor to hypertension. Variable functional responses to U-II, both within and between species studied to date, complicate the characterization of UT antagonists. In the cat, however, U-II causes systemic hypertension and constricts arterial segments isolated from several vascular beds. The purpose of this study was to clone and pharmacologically characterize cat recombinant UT to determine whether this system represents a model for characterizing UT antagonists. Cloned cat UT displayed 74% identity to primate UT, and 77% identity to rodent UT. [(125)I] hU-II bound in a saturable manner to a single site on recombinant cat UT with high affinity (K(D) 288+/-13pM) and high density (B(max) 747+/-66fmol/mg protein). U-II isopeptides displayed equipotent, high affinity binding to cat UT (K(i) 1.8-5.3nM). Cat UT was coupled to intracellular [Ca(2+)] release (EC(50) 0.6+/-0.2nM) and total inositol phosphate (IP) formation (EC(50) 0.4+/-0.1nM). Protein kinase C activation desensitized cat, but not human, UT-mediated IP formation. UT mRNA expression was detected in cat blood vessels, trachea, lung, and kidney, where the medulla (K(D) 815+/-34) and cortex and (K(D) 316+/-39pM) displayed high affinity binding for human U-II (hU-II). The cat urotensin-II receptor represents a suitable in vitro model to examine the role of the U-II/UT system in the etiology of hypertension, assisting in the evaluation of the UT antagonists to help treat cardiovascular disease.


Subject(s)
Cloning, Molecular , Receptors, G-Protein-Coupled/genetics , Amino Acid Sequence , Animals , Calcium Signaling/physiology , Cats , Cell Line , Cell Membrane/physiology , Conserved Sequence , DNA Primers , Humans , Inositol Phosphates/metabolism , Mice , Molecular Sequence Data , Organ Specificity , Polymerase Chain Reaction , RNA, Messenger/genetics , Rats , Receptors, G-Protein-Coupled/drug effects , Receptors, G-Protein-Coupled/metabolism , Recombinant Proteins/drug effects , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Transfection
9.
J Immunol ; 173(12): 7230-8, 2004 Dec 15.
Article in English | MEDLINE | ID: mdl-15585845

ABSTRACT

Neuromedin U (NmU), originally isolated from porcine spinal cord and later from other species, is a novel peptide that potently contracts smooth muscle. NmU interacts with two G protein-coupled receptors designated as NmU-1R and NmU-2R. This study demonstrates a potential proinflammatory role for NmU. In a mouse Th2 cell line (D10.G4.1), a single class of high affinity saturable binding sites for (125)I-labeled NmU (K(D) 364 pM and B(max) 1114 fmol/mg protein) was identified, and mRNA encoding NmU-1R, but not NmU-2R, was present. Competition binding analysis revealed equipotent, high affinity binding of NmU isopeptides to membranes prepared from D10.G4.1 cells. Exposure of these cells to NmU isopeptides resulted in an increase in intracellular Ca(2+) concentration (EC(50) 4.8 nM for human NmU). In addition, NmU also significantly increased the synthesis and release of cytokines including IL-4, IL-5, IL-6, IL-10, and IL-13. Studies using pharmacological inhibitors indicated that maximal NmU-evoked cytokine release required functional phospholipase C, calcineurin, MEK, and PI3K pathways. These data suggest a role for NmU in inflammation by stimulating cytokine production by T cells.


Subject(s)
Cytokines/metabolism , Membrane Proteins/physiology , Neuropeptides/physiology , Receptors, Neurotransmitter/physiology , Th2 Cells/immunology , Th2 Cells/metabolism , Animals , Calcineurin/physiology , Calcium/metabolism , Cell Line , Clone Cells , Cytokines/antagonists & inhibitors , Dogs , Estrenes/pharmacology , Humans , Interleukins/antagonists & inhibitors , Interleukins/metabolism , MAP Kinase Signaling System/immunology , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Mice , Mitogen-Activated Protein Kinases/metabolism , Neuropeptides/metabolism , Phosphatidylinositol 3-Kinases/physiology , Pyrrolidinones/pharmacology , Rats , Receptors, Interleukin-4/physiology , Receptors, Neurotransmitter/biosynthesis , Receptors, Neurotransmitter/genetics , Signal Transduction/immunology , Swine , Type C Phospholipases/antagonists & inhibitors , Type C Phospholipases/physiology
10.
Peptides ; 25(8): 1339-47, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15350702

ABSTRACT

Plasma and urinary levels of "urotensin(U)-II-like" substances determined in healthy human volunteers were 12.4 +/- 0.6 ng/ml and 2.2 +/- 0.3 ng/ml by RIA, an order of magnitude lower than that seen by RRA, 167.5 +/- 9.5 ng/ml and 65.2 +/- 4.3 ng/ml. HPLC demonstrated the existence of at least three prominent activity peaks in plasma and urine, the more hydrophobic of which did not co-elute with U-II, degradation products or URP. RRA and RIA recognized these peaks with contrasting efficacy. As such, published levels of "U-II-like" activity should be interpreted with caution until a better understanding is obtained regarding what species specific RIA and RRA assay reagents interact with.


Subject(s)
Radioligand Assay/methods , Urotensins , Adult , Female , Humans , Male , Middle Aged , Radioimmunoassay/methods , Reference Values , Reproducibility of Results , Urotensins/analysis , Urotensins/blood , Urotensins/urine
11.
Pharmacology ; 72(1): 33-41, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15292653

ABSTRACT

Neuromedin U (NmU) is a smooth muscle contracting peptide. Recently, two G-protein-coupled receptors for NmU (NmU1R and NmU2R) have been cloned having approximately 50% homology. They have distinct patterns of expression suggesting they may have different biological functions. This study provides a comprehensive characterization of both NmU receptors expressed in human embryonic kidney 293 cells. [125I]hNmU binding to the recombinant NmU receptors was rapid, saturable, of high affinity and to a single population of binding sites. Exposure of these cells to NmU isopeptides resulted in an increase in intracellular [Ca2+]i release (EC50 value of 0.50 +/- 0.10 nmol/l) and inositol phosphate formation (EC50 1.6 +/- 0.2 and 1.50 +/- 0.4 nmol/l for NmU1R and NmU2R respectively). Furthermore, hNmU inhibited forskolin (3 micromol/l)-stimulated accumulation of cAMP in intact HEK-293 cells expressing either NmU1R or NmU2R. The inhibitory effect was significant for the cells expressing NmU2R with IC50 value of 0.80 +/- 0.21 nmol/l. In summary, both NmU1R and NmU2R in HEK-293 cells have similar signaling capability.


Subject(s)
Membrane Proteins/metabolism , Receptors, Neurotransmitter/metabolism , Binding Sites , Cells, Cultured , Clone Cells , Humans , Kidney/metabolism , Membrane Proteins/genetics , Membrane Proteins/physiology , Oligonucleotide Array Sequence Analysis , Radioligand Assay , Receptors, Neurotransmitter/genetics , Receptors, Neurotransmitter/physiology , Signal Transduction
12.
Br J Pharmacol ; 142(6): 921-32, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15210573

ABSTRACT

1 In an effort to identify endogenous, native mammalian urotensin-II (U-II) receptors (UT), a diverse range of human, primate and rodent cell lines (49 in total) were screened for the presence of detectable [125I]hU-II binding sites. 2 UT mRNA (Northern blot, PCR) and protein (immunocytochemistry) were evident in human skeletal muscle tissue and cells. 3 [(125)I]hU-II bound to a homogenous population of high-affinity, saturable (Kd 67.0+/-11.8 pm, Bmax 9687+/-843 sites cell(-1)) receptors in the skeletal muscle (rhabdomyosarcoma) cell line SJRH30. Radiolabel was characteristically slow to dissociate (< or =15% dissociation 90 min). A lower density of high-affinity U-II binding sites was also evident in the rhabdomyosarcoma cell line TE671 (1667+/-165 sites cell(-1), Kd 74+/-8 pm). 4 Consistent with the profile recorded in human recombinant UT-HEK293 cells, [125I]hU-II binding to SJRH30 cells was selectively displaced by both mammalian and fish U-II isopeptides (Kis 0.5+/-0.1-1.2+/-0.3 nm) and related analogues (hU-II[4-11]>[Cys(5,10)]Acm hU-II; Kis 0.4+/-0.1 and 864+/-193 nm, respectively). 5 U-II receptor activation was functionally coupled to phospholipase C-mediated [Ca2+]i mobilization (EC50 6.9+/-2.2 nm) in SJRH30 cells. 6 The present study is the first to identify the presence of 'endogenous' U-II receptors in SJRH30 and TE671 cells. SJRH30 cells, in particular, might prove to be of utility for (a) investigating the pharmacological properties of hU-II and related small molecule antagonists at native human UT and (b) delineating the role of this neuropeptide in the (patho)physiological regulation of mammalian neuromuscular function.


Subject(s)
Receptors, G-Protein-Coupled/metabolism , Rhabdomyosarcoma/metabolism , Animals , Binding Sites , Binding, Competitive/drug effects , Blotting, Northern , Calcitonin Gene-Related Peptide/pharmacology , Calcium/metabolism , Cell Line , Cell Line, Tumor , Dose-Response Relationship, Drug , Female , Gene Expression Profiling , HeLa Cells , Humans , Hypothalamic Hormones/pharmacology , Immunohistochemistry , Intracellular Space/drug effects , Intracellular Space/metabolism , Iodine Radioisotopes , Kinetics , Male , Melanins/pharmacology , Neuropeptide Y/pharmacology , Neurophysins/pharmacology , Pertussis Toxin/pharmacology , Pituitary Hormones/pharmacology , Protein Precursors/pharmacology , Radioligand Assay , Reverse Transcriptase Polymerase Chain Reaction , Rhabdomyosarcoma/pathology , Thapsigargin/pharmacology , Urotensins/genetics , Urotensins/metabolism , Urotensins/pharmacology , Vasopressins/pharmacology
13.
Biochem Pharmacol ; 63(11): 1949-59, 2002 Jun 01.
Article in English | MEDLINE | ID: mdl-12093471

ABSTRACT

A complementary DNA encoding calcitonin receptor-like receptor (CRLR) was isolated from a bovine aortic endothelial cell library. The bovine CRLR has 462 amino acids and 92% homology with the human CRLR. In a reverse transcriptase-polymerase chain reaction assay, bovine CRLR was found to be widely distributed, including in the heart and lungs. Stable transfection of bovine CRLR in human embryonic kidney cells (HEK-293) resulted in specific high-affinity [125I] rat adrenomedulin (rADM)-binding (dissociation constant=145+/-15 pM). ADM-stimulated adenylyl cyclase activity with an EC50 value of 5.0+/-1.2 nM. The human ADM receptor antagonist hADM(22-52) inhibited [125I]rADM-binding and ADM-stimulated adenylyl cyclase activity. Interactions between bovine CRLR and individual receptor activity modifying proteins (RAMPs) were also investigated. Transient co-transfection of bovine CRLR cDNA with human receptor activity modifying protein 1 (hRAMP1) cDNA in HEK-293 cells resulted in the expression of a CRLR that displayed high-affinity binding to calcitonin gene-related peptide. Co-transfection of bovine CRLR with human RAMP2 or RAMP3 cDNAs in HEK-293 cells displayed high-affinity ADM receptors. These observations suggest that in the absence of exogenous RAMPs heterologous expression of bovine CRLR results in an ADM receptor phenotype.


Subject(s)
Endothelium, Vascular/metabolism , Receptors, Calcitonin/genetics , Receptors, Peptide/metabolism , Adrenomedullin , Amino Acid Sequence , Animals , Aorta/cytology , Calcitonin Receptor-Like Protein , Cattle , Cells, Cultured , Cloning, Molecular , Endothelium, Vascular/drug effects , Humans , Intracellular Signaling Peptides and Proteins , Membrane Proteins/metabolism , Molecular Sequence Data , Peptides/pharmacology , Receptor Activity-Modifying Protein 2 , Receptor Activity-Modifying Protein 3 , Receptor Activity-Modifying Proteins , Receptors, Adrenomedullin , Receptors, Calcitonin/metabolism , Receptors, Peptide/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid
14.
J Pharmacol Exp Ther ; 301(3): 987-92, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12023529

ABSTRACT

Two endogenous receptors for the potent smooth muscle-stimulating peptide neuromedin U (NmU) have recently been identified and cloned. Pharmacological, binding, and expression studies were conducted in an attempt to determine the receptor(s) involved in the smooth muscle-stimulating effects of NmU. The NmU peptides caused a concentration-dependent contraction of canine isolated urinary bladder. NmU did not have this same effect in the urinary bladder from rat, guinea pig, rabbit, mouse, or ferret. Although NmU had no effect on canine uterus it did cause contraction of canine stomach, ileum, and colon. As well as causing contraction of canine bladder in vitro, NmU administered systemically resulted in a significant increase in urinary bladder pressure in vivo. High-affinity binding sites for NmU were identified in canine bladder. The four NmU peptides porcine NmU-8, rat NmU-23, human NmU-25, and porcine NmU-25 displaced (125)I-NmU-25 binding with similar K(i) values (0.08-0.24 nM). A different binding profile was revealed in human embryonic kidney-293 cells transiently expressed with the canine NmU-2 receptor where porcine NmU-8 (K(i) = 147.06 nM) was much less potent than the other NmU peptides. Using TaqMan, expression of NmU-1 was detected in human urinary bladder, small intestine, colon, and uterus. Expression of NmU-2 was much lower or absent in these human tissues and undetectable in canine bladder and stomach. The results of this study reveal significant species differences in the activity of NmU. The contractile activity in human and canine smooth muscle seems to be mediated by the recently cloned NmU-1 receptor.


Subject(s)
Membrane Proteins , Muscle, Smooth/drug effects , Muscle, Smooth/metabolism , Neuropeptides/metabolism , Receptors, Neurotransmitter , Animals , Binding Sites/drug effects , Binding Sites/physiology , Cell Line , Dogs , Drug Evaluation, Preclinical/methods , Female , Ferrets , Guinea Pigs , Humans , Intestinal Mucosa/metabolism , Intestines/drug effects , Male , Mice , Muscle Contraction/drug effects , Muscle Contraction/physiology , Neuropeptides/pharmacology , Rabbits , Rats , Receptors, Cell Surface/biosynthesis , Urinary Bladder/drug effects , Urinary Bladder/metabolism , Uterus/drug effects , Uterus/metabolism
15.
Br J Pharmacol ; 136(1): 9-22, 2002 May.
Article in English | MEDLINE | ID: mdl-11976263

ABSTRACT

Urotensin-II (U-II) and its receptor (UT) represent novel therapeutic targets for management of a variety of cardiovascular diseases. To test such hypothesis, it will be necessary to develop experimental animal models for the manipulation of U-II/UT receptor system. The goal of this study was to clone mouse and primate preproU-II and UT for pharmacological profiling. Monkey and mouse preproU-II genes were identified to encode 123 and 125 amino acids. Monkey and mouse UT receptors were 389, and 386 amino acids, respectively. Genomic organization of mouse genes showed that the preproU-II has four exons, while the UT receptor has one exon. Although initially viewed by many exclusively as cardiovascular targets, the present study demonstrates expression of mouse and monkey U-II/UT receptor mRNA in extra-vascular tissue including lung, pancreas, skeletal muscle, kidney and liver. Ligand binding studies showed that [125I]h U-II bound to a single sites to the cloned receptors in a saturable/high affinity manner (Kd 654+/-154 and 214+/-65 pM and Bmax of 1011+/-125 and 497+/-68 fmol mg-1 for mouse and monkey UT receptors, respectively). Competition binding analysis demonstrated equipotent, high affinity binding of numerous mammalian, amphibian and piscine U-II isopeptides to these receptors (Ki=0.8 - 3 nM). Fluorescein isothiocyanate (FITC) labelled U-II, bound specifically to HEK-293 cells expressing mouse or monkey UT receptor, confirming cell surface expression of recombinant UT receptor. Exposure of these cells to human U-II resulted in an increase in intracellular [Ca2+] concentrations (EC50 3.2+/-0.8 and 1.1+/-0.3 nM for mouse and monkey UT receptors, respectively) and inositol phosphate (Ip) formation (EC50 7.2+/-1.8 and 0.9+/-0.2 nM for mouse and monkey UT receptors, respectively) consistent with the primary signalling pathway for UT receptor involving phospholipase C activation.


Subject(s)
GTP-Binding Proteins/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Receptors, G-Protein-Coupled , Urotensins/genetics , Urotensins/metabolism , Amino Acid Sequence , Animals , Binding Sites , Binding, Competitive , Blotting, Southern , Cell Line , Cloning, Molecular , Cricetinae , Fluorescein-5-isothiocyanate , Fluorescent Dyes , Humans , Macaca fascicularis , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Organ Specificity , Protein Precursors/genetics , Protein Precursors/metabolism , Radioligand Assay , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...