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










Publication year range
1.
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
2.
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
3.
J Pharmacol Exp Ther ; 295(1): 139-45, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10991971

ABSTRACT

STI571 (formerly known as CGP 57148B) is a protein-tyrosine kinase inhibitor that is currently in clinical trials for the treatment of chronic myelogenous leukemia. STI571 selectively inhibits the Abl and platelet-derived growth factor (PDGF) receptor tyrosine kinases in vitro and blocks cellular proliferation and tumor growth of Bcr-abl- or v-abl-expressing cells. We have further investigated the profile of STI571 against related receptor tyrosine kinases. STI571 was found to potently inhibit the kinase activity of the alpha- and beta-PDGF receptors and the receptor for stem cell factor, but not the closely related c-Fms, Flt-3, Kdr, Flt-1, and Tek tyrosine kinases. Additionally, no inhibition of c-Met or nonreceptor tyrosine kinases such as Src and Jak-2 has been observed. In cell-based assays, STI571 selectively inhibited PDGF and stem cell factor-mediated cellular signaling, including ligand-stimulated receptor autophosphorylation, inositol phosphate formation, and mitogen-activated protein kinase activation and proliferation. These results expand the profile of STI571 and suggest that in addition to chronic myelogenous leukemia, STI571 may have clinical potential in the treatment of diseases that involve abnormal activation of c-Kit or PDGF receptor tyrosine kinases.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Piperazines/pharmacology , Proto-Oncogene Proteins c-abl/antagonists & inhibitors , Proto-Oncogene Proteins c-kit/drug effects , Pyrimidines/pharmacology , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , Signal Transduction/drug effects , Animals , Benzamides , Cell Line , Imatinib Mesylate , Mice , Mitogen-Activated Protein Kinases/physiology , Proto-Oncogene Proteins c-kit/physiology , Receptors, Platelet-Derived Growth Factor/physiology , Stem Cell Factor/physiology
4.
Biochem Pharmacol ; 59(9): 1033-43, 2000 May 01.
Article in English | MEDLINE | ID: mdl-10704932

ABSTRACT

17-fold) increase in DNA fragmentation. Fluorescence microscopy, using DNA binding dyes, demonstrated that cell death following hypoxia/reoxygenation was due predominantly to apoptosis and not necrosis. Furthermore, reoxygenation, but not hypoxia alone, caused a time-dependent increase in the activation of JNK as monitored by western blot analysis using a phospho-specific JNK antibody. In contrast, p38 mitogen-activated protein kinase was activated following hypoxia, but this activation was not augmented during reoxygenation. Exposure of human kidney cells to a 2'-methoxyethyl mixed backbone antisense oligonucleotide directed against human JNK1 (JNK1 AS) resulted in a potent suppression of JNK mRNA and protein expression, whereas a 6-base mismatch control oligonucleotide was without effect. Moreover, a significant diminution of reoxygenation-induced apoptosis was observed in cells exposed to JNK1 AS but not to the mismatch control oligonucleotide. Taken together, these results strongly indicate that activation of the JNK signaling cascade is a major mechanism whereby hypoxia/reoxygenation induces apoptosis.


Subject(s)
Kidney/drug effects , Mitogen-Activated Protein Kinases/metabolism , Oligonucleotides, Antisense/pharmacology , Oxygen/physiology , Apoptosis , Calcium-Calmodulin-Dependent Protein Kinases/biosynthesis , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Division/drug effects , Cell Hypoxia/physiology , Cells, Cultured , Enzyme Activation , Humans , JNK Mitogen-Activated Protein Kinases , Kidney/cytology , Kidney/enzymology , Mitogen-Activated Protein Kinases/biosynthesis , Mitogen-Activated Protein Kinases/genetics , Necrosis , p38 Mitogen-Activated Protein Kinases
5.
Bioorg Med Chem Lett ; 10(2): 183-7, 2000 Jan 17.
Article in English | MEDLINE | ID: mdl-10673107

ABSTRACT

Opioid binding affinities were assessed for a series of cyclazocine analogues where the prototypic 8-OH substituent of cyclazocine was replaced by amino and substituted-amino groups. For mu and kappa opioid receptors, secondary amine derivatives having the (2R,6R,11R)-configuration had the highest affinity. Most targets were efficiently synthesized from the triflate of cyclazocine or its enantiomers using Pd-catalyzed amination procedures.


Subject(s)
Analgesics/chemical synthesis , Benzeneacetamides , Cyclazocine/analogs & derivatives , Analgesics/pharmacology , Animals , Binding, Competitive , Brain/drug effects , Cyclazocine/chemistry , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/metabolism , Guinea Pigs , Molecular Structure , Naltrexone/analogs & derivatives , Naltrexone/metabolism , Narcotic Antagonists , Pyrrolidines/metabolism , Receptors, Opioid/agonists , Stereoisomerism , Structure-Activity Relationship
7.
Mol Pharmacol ; 53(1): 97-104, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9443936

ABSTRACT

To characterize the pharmacodynamic properties of CGP 69846A/ISIS 5132, an antisense oligodeoxynucleotide directed against the mitogenic signal transducer Raf-1 kinase, we investigated the elicited biological responses in human coronary artery smooth muscle cells. Cell exposure to CGP 69846A resulted in a reversible time- and concentration-dependent down-regulation of cellular Raf-1 gene expression and, ultimately, inhibition of cell cycle progression. The highest potencies of this compound to reduce Raf-1 mRNA and protein levels were observed after 24 and 48 hr of cell exposure, respectively, with corresponding IC50 values of approximately 100 and approximately 300 nM. Proliferation was inhibited with an IC50 value of approximately 300 nM after 72 hr. We interpreted the recovery rate of Raf-1 mRNA after cell exposure to antisense ODNs as the half-life (t1/2 approximately 50 hr) of active intracellular CGP 69846A in our cell culture system. The endogenous Raf-1 turnover half-life of approximately 30 hr, as assessed by monitoring metabolically labeled Raf-1 protein, correlated kinetically with the antisense-induced protein decay rate (50% decay in approximately 33 hr), indicating that the efficiency of CGP 69846A in decreasing Raf-1 protein levels was rate-limited by the endogenous protein turnover rate. The pharmacodynamic effects of CGP 69846A antisense ODNs are therefore limited by the duration of its intracellular activity rather than by its ability to transiently decrease mRNA levels. Local steady state exposure to CGP 69846A may represent a new approach to prevent the transition of quiescent vascular smooth muscle cells into the pathologically hyperproliferating cells seen after angioplasty.


Subject(s)
Antineoplastic Agents/pharmacology , Muscle, Smooth, Vascular/drug effects , Oligodeoxyribonucleotides, Antisense , Oligonucleotides, Antisense/pharmacology , Proto-Oncogene Proteins c-raf/genetics , Thionucleotides/pharmacology , Cell Cycle/drug effects , Cell Division/drug effects , Cells, Cultured , Coronary Vessels/cytology , Coronary Vessels/drug effects , Coronary Vessels/metabolism , Humans , Kinetics , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Proto-Oncogene Proteins c-raf/biosynthesis , Proto-Oncogene Proteins c-raf/metabolism , RNA, Messenger/metabolism , Transcription, Genetic/drug effects
8.
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
9.
Biochem Biophys Res Commun ; 227(1): 176-81, 1996 Oct 03.
Article in English | MEDLINE | ID: mdl-8858121

ABSTRACT

A low molecular weight endothelin (ET-1) inhibitor (Ex. 127, European Patent Application 404 525 A2, Takeda Chemical Ind., 1991), CGS 26061, was synthesized and evaluated to determine its mechanism of action. CGS 26061 (10 microM) failed to inhibit binding of [125I]ET-1 in porcine thoracic aorta and was without effect on ET-1-induced [3H]inositol phosphate accumulation in A7r5 cells. However, CGS 26061 relaxed porcine coronary arterial rings precontracted with ET-1. In addition, contractions to PGF2 alpha and low K+ (20 mM) but not high K+ were attenuated, suggesting that CGS 26061 (1, 10 microM) is a potassium channel opener. Patch-clamp experiments confirmed the K+ channel activity (0.1-10 microM). The originally re ported inhibition of ET-1-induced pressor responses by Ex. 127 (CGS 26061) was not replicated in the anesthetized dog or conscious rat nor was it shown to be antihypertensive in SHR. These data have identified CGS 26061 as a novel K+ channel opener with a unique cardiovascular profile.


Subject(s)
Endothelin-1/antagonists & inhibitors , Potassium Channels/drug effects , Pyrimidinones/pharmacology , Thiazines/pharmacology , Vasodilator Agents/pharmacology , Animals , Blood Pressure/drug effects , Dogs , In Vitro Techniques , Male , Rats , Rats, Inbred SHR , Rats, Sprague-Dawley , Species Specificity , Swine
10.
J Pharmacol Exp Ther ; 276(1): 74-83, 1996 Jan.
Article in English | MEDLINE | ID: mdl-8558459

ABSTRACT

CGS 27830 [meso-1,4-dihydro-5-methoxycarbonyl-2,6-dimethyl-4-(3- nitrophenyl)-3-pyridine carboxylic acid anhydride] is a nonpeptidic, insurmountable, endothelin (ET) receptor antagonist with approximately 10- to 20-fold selectivity for ETA receptors. CGS 27830 exhibits unusual binding properties which depend on the receptor and ligand: standard saturation binding experiments (coincubation of membranes with ligand in the absence or presence of antagonist) suggest that CGS 27830 is a competitive inhibitor of [125I]IRL 1620 binding to ETB receptors in rat cerebellar membranes (i.e., there was a change of apparent Kd with no change of maximum binding), but a noncompetitive inhibitor of [125I]IRL 1620 binding to ETB receptors in rat lung membranes (i.e., significant loss of total binding was observed). Although the antagonist appears to be a noncompetitive inhibitor of [125I]IRL 1620 binding to ETB receptors in rat lung membranes, CGS 27830 appears to be a competitive inhibitor of [125I]ET-1 binding to the same receptors as well as to ETA receptors in A7r5 cell membranes. Thus, CGS 27830 can distinguish [125I]IRL 1620 binding to ETB receptors in rat cerebellar and lung membranes, but not ET-1 binding to ETB receptors in these tissues. These unusual binding properties demonstrate that rat lung and cerebellum ETB receptors interact differently with IRL 1620 or ET-1.


Subject(s)
Dihydropyridines/pharmacology , Endothelin Receptor Antagonists , Receptors, Endothelin/agonists , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/physiology , Cell Membrane/drug effects , Cell Membrane/ultrastructure , Cells, Cultured , Cerebellum/drug effects , Cerebellum/metabolism , Cerebellum/ultrastructure , Endothelins/metabolism , In Vitro Techniques , Iodine Radioisotopes , Kinetics , Ligands , Lung/drug effects , Lung/metabolism , Lung/ultrastructure , Male , Muscle Contraction/drug effects , Peptide Fragments/metabolism , Protein Binding/drug effects , Radioligand Assay , Rats , Rats, Sprague-Dawley , Receptors, Endothelin/metabolism
11.
J Cardiovasc Pharmacol ; 22 Suppl 8: S168-70, 1993.
Article in English | MEDLINE | ID: mdl-7509934

ABSTRACT

In this study, the short-term regulation of endothelin-1-(ET-1) induced phosphoinositide (PI) hydrolysis and arachidonic acid release were investigated in cultured rat aortic smooth-muscle cells. ET-1, but not the ETB-selective peptide sarafotoxin (SFX) S6c, induced a dose-dependent increase in [3H]inositol phosphate release (EC50 = 0.4 +/- 0.1 nM). ET-3 stimulated this response only at concentrations > 0.1 microM. The ETA receptor antagonist BQ-123 inhibited ET-1-induced PI turnover, with an IC50 value of 97 +/- 15 nM. Pre-exposure of intact cells to ET-1 resulted in a 72% and 73% reduction in the ability of ET-1 or SFX S6b, respectively, to stimulate [3H]inositol phosphate release, without affecting the response to vasopressin. In contrast, PI turnover induced by ET-1 or SFX S6b was only slightly lowered, by 28% and 22%, after a 30-min preincubation period with SFX S6b. ET-1, but not SFX S6c, also stimulated [3H]arachidonic acid release by two-fold (EC50 = 3 +/- 0.8 nM). Pretreatment of intact cells with neomycin or phorbol-12-myristate-13-acetate resulted in a 49% and 44% inhibition of ET-1-induced [3H]inositol phosphate accumulation but did not decrease ET-1-stimulated [3H]arachidonic acid release, suggesting that these responses are separately regulated events in these cells.


Subject(s)
Arachidonic Acids/metabolism , Muscle, Smooth, Vascular/metabolism , Phosphatidylinositols/metabolism , Receptors, Endothelin/metabolism , Animals , Cells, Cultured , Endothelins/pharmacology , Hydrolysis , Muscle, Smooth, Vascular/drug effects , Rats , Vasodilator Agents/pharmacology , Viper Venoms/pharmacology
12.
J Pharmacol Exp Ther ; 262(2): 611-8, 1992 Aug.
Article in English | MEDLINE | ID: mdl-1323655

ABSTRACT

The ability of endothelin (ET) isopeptides to interact with ET receptor subtypes and stimulate phosphoinositide (PI) hydrolysis was examined in the rat lung. [125I]ET-1 and [125I]ET-3 binding to lung homogenates was saturable with maximal binding capacity values of 438 and 125 fmol/mg of protein and Kd values of 29 and 13 pM. The nonselective peptides, ET-1 and ET-2, produced steep inhibition of both [125I]ET-1 and [125I] ET-3 binding. The ETB-selective peptides, ET-3, sarafotoxin (SFX) S6a, SFX S6b and SFX S6c and the ETA-selective antagonist, BQ-123, generated shallow inhibition curves of [125I]ET-1 binding indicating the presence of both ETA and ETB receptors in the lung. Whereas the peptides exhibited similar potency in stimulating PI turnover in rat lung slices, the ability of ET-3 (1.6-fold) and SFX S6c (2-fold) to maximally stimulate [3H]inositol phosphate release was significantly different from the maximal response produced by ET-1 (4-fold) or SFX S6b (3.2-fold). The ETA-selective antagonist, BQ-123 [cyclo(L-Leu-D-Trp-D-Asp-L-Pro-D-Val)], inhibited PI hydrolysis induced by ET-1 or SFX S6b by approximately 80%, although having no effect on ET-3- or SFX S6c-induced PI turnover. Furthermore, ET-1- and SFX S6b-stimulated [3H]inositol phosphate release was significantly decreased in the presence of quinacrine and nordihydroguairetic acid, but not indomethacin. In contrast, these inhibitors had no effect on PI hydrolysis induced by SFX S6c.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Endothelins/pharmacology , Lung/metabolism , Phosphatidylinositols/metabolism , Receptors, Cell Surface/drug effects , Animals , Arachidonic Acid/metabolism , Endothelins/metabolism , Hydrolysis , Male , Peptides, Cyclic/pharmacology , Rats , Rats, Inbred Strains , Receptors, Cell Surface/analysis , Receptors, Endothelin
13.
J Neurochem ; 54(5): 1725-34, 1990 May.
Article in English | MEDLINE | ID: mdl-2157816

ABSTRACT

The actions of tumor promoters on the coupling of muscarinic receptors to the hydrolysis of inositol lipids and the generation of Ca2+ signals were examined in the human neuroblastoma SH-SY5Y cell line. Pretreatment of SH-SY5Y cells with 50 nM 12-O-tetradecanoylphorbol 13-acetate (TPA) for 5 days resulted in neuronal differentiation, a 28% decrease in both N-[3H]methylscopolamine and [3H]-scopolamine binding, and a significantly larger reduction (48%) in agonist-stimulated 3H-inositol phosphate generation. Whereas mezerein could mimic the effects produced by TPA, the biologically inactive 4 alpha-phorbol 12,13-didecanoate was without effect on both antagonist binding and agonist-stimulated phosphoinositide (PPI) turnover. A decline (approximately 50%) in the agonist-mediated rise in cytoplasmic Ca2+ and a substantial loss of protein kinase C activity also were observed following pretreatment with TPA or mezerein. The ability of fluoride, an agent capable of direct activation of guanine nucleotide binding proteins, to stimulate 3H-inositol phosphate release was significantly reduced in SH-SY5Y cells treated with these agents. Furthermore, pretreatment of SH-SY5Y neuroblastoma cells with TPA or mezerein impaired 3H-inositol phosphate formation induced by the addition of either guanosine 5'-O-(3-thiotriphosphate) or carbamylcholine to digitonin-permeabilized cells, but not that elicited by the addition of 2 mM CaCl2. Although cells cultured in the presence of serum-free media also exhibited neuronal differentiation, no significant alteration in either muscarinic receptor number or agonist-stimulated PPI hydrolysis was observed. The results suggest that TPA and mezerein decrease agonist-stimulated PPI hydrolysis and Ca2+ signaling in SH-SY5Y cells not only by a reduction in muscarinic receptor number but also through an inhibition of guanine nucleotide-stimulated PPI turnover.


Subject(s)
Diterpenes , Guanine Nucleotides/pharmacology , Neuroblastoma/metabolism , Phosphatidylinositols/metabolism , Receptors, Muscarinic/metabolism , Calcium/metabolism , Culture Media , Cytoplasm/metabolism , Guanosine 5'-O-(3-Thiotriphosphate) , Guanosine Triphosphate/analogs & derivatives , Guanosine Triphosphate/pharmacology , Humans , Hydrolysis , Inositol Phosphates/metabolism , Neuroblastoma/pathology , Osmolar Concentration , Protein Kinase C/metabolism , Sodium Fluoride/pharmacology , Terpenes/pharmacology , Tetradecanoylphorbol Acetate/pharmacology , Thionucleotides/pharmacology , Time Factors , Tumor Cells, Cultured
14.
Membr Biochem ; 9(1): 9-27, 1990.
Article in English | MEDLINE | ID: mdl-2077364

ABSTRACT

Like other neurotransmitter receptors, muscarinic acetylcholine receptors are subject to regulation by the state of receptor activation. Prolonged increases in the concentration of muscarinic agonists result in a decrease in receptor density and loss of receptor sensitivity, both in vivo and in vitro. On the other hand, when the receptor is deprived of acetylcholine for a long duration in vivo, the receptor becomes more sensitive in responding to muscarinic agonists. However, it has been more difficult to demonstrate increases in receptor concentration that accompany this supersensitive state. The purpose of this review is to provide current information related to the characteristics of muscarinic receptor regulation and the molecular mechanisms underlying this phenomenon, regarding both the density of receptors and their transduction mechanisms. Furthermore, possible feedback regulatory roles of different second messenger signals are discussed. Particular emphasis is dedicated to molecular mechanisms of regulation of neuronal muscarinic receptors.


Subject(s)
Neurons/metabolism , Receptors, Muscarinic/metabolism , Animals , Cells, Cultured , Humans , Parasympatholytics/pharmacology , Parasympathomimetics/pharmacology
15.
Biochem Pharmacol ; 38(11): 1827-34, 1989 Jun 01.
Article in English | MEDLINE | ID: mdl-2544180

ABSTRACT

Short-term agonist-induced loss of cell surface muscarinic receptors and desensitization of receptor-mediated cyclic GMP (cGMP) formation and phosphoinositide hydrolysis were examined in mouse neuroblastoma cells (clone N1E-115) in suspension. This treatment resulted in a time-dependent reduction of approximately 40% of the specific binding of the hydrophilic antagonist [3H]N-methyl-scopolamine [( 3 H]NMS) with a T 1/2 of down-regulation of 4.83 min. Scatchard analysis revealed that brief exposure to the agonist resulted in a significant reduction in the Bmax with no change in the Kd. Agonist-induced cGMP formation decreased in a similar time-dependent manner with an average T 1/2 of 4.79 min. However, desensitization of muscarinic receptor-stimulated accumulation of inositol phosphates demonstrated a much slower time-course and was accompanied by a reduction in the maximal response with no change in the EC50. In addition, there was rapid partial recovery of cell surface receptors and desensitized cGMP response, with no apparent resensitization of phosphoinositide hydrolysis. Thus, there was a differential rate of short-term desensitization and resensitization of these two muscarinic receptor-mediated responses. Moreover, desensitization of cGMP formation, but not phosphoinositide hydrolysis, closely paralleled loss of cell surface muscarinic receptors.


Subject(s)
Cyclic GMP/physiology , Neuroblastoma/physiopathology , Phosphatidylinositols/physiology , Receptors, Muscarinic/drug effects , Animals , Carbachol/pharmacology , In Vitro Techniques , Inositol Phosphates/metabolism , Mice , N-Methylscopolamine , Scopolamine Derivatives/pharmacology , Time Factors
16.
J Pharmacol Exp Ther ; 247(3): 934-40, 1988 Dec.
Article in English | MEDLINE | ID: mdl-2849677

ABSTRACT

Pirenzepine selectively antagonized muscarinic receptor-mediated cyclic GMP formation in a noncompetitive fashion in mouse neuroblastoma cells (clone N1E-115). These effects of pirenzepine were time- and concentration-dependent and they were also reversible. Interestingly, whereas atropine elicited competitive antagonism of the cyclic GMP response at low concentrations, it also behaved like a noncompetitive antagonist at higher concentrations and its effects were partially reversible. Using additional approaches to study the mechanisms underlying this anomalous antagonistic profile of pirenzepine, we investigated whether this deviation from competition could be due to the short time of exposure to muscarinic agonists (30 sec) used in cyclic GMP measurements. Our data indicated that the mode of pirenzepine-induced antagonism of ligand binding to muscarinic receptors was different when assessed using nonequilibrium (30 sec) or equilibrium (1 hr) incubations. Thus, pirenzepine appeared to be noncompetitive and competitive under these two conditions, respectively. Furthermore, although pirenzepine blocked receptor-mediated phosphoinositide hydrolysis competitively when the response was measured at 20 min, it was clearly noncompetitive using 5-min incubations. Therefore, the noncompetitive antagonism by pirenzepine detected in cyclic GMP measurements might be only apparent and might be attributed, at least in part, to a lack of an equilibrium state under the specific conditions of these assays.


Subject(s)
Cyclic GMP/biosynthesis , Phosphatidylinositols/metabolism , Pirenzepine/pharmacology , Receptors, Muscarinic/drug effects , Animals , Atropine/pharmacology , Carbachol/pharmacology , Hydrolysis , Mice , N-Methylscopolamine , Scopolamine Derivatives/metabolism
17.
Eur J Pharmacol ; 156(1): 35-45, 1988 Oct 26.
Article in English | MEDLINE | ID: mdl-2850209

ABSTRACT

There was a 25 and 27% reduction in the density of mouse brain muscarinic acetylcholine receptors 18 and 24 h following a single injection of the organophosphate diisopropylfluorophosphate (DFP) when the muscarinic antagonist [3H]N-methylscopolamine ([3H]NMS) was used as the ligand. Down-regulation of specific [3H]NMS binding was rapidly reversible reaching control levels 36 h after DFP administration. Carbamylcholine and pirenzepine competition for the specific binding of either [3H]NMS or [3H]quinuclidinyl benzilate ([3H]QNB) in brain homogenates from untreated and DFP-treated mice demonstrated that the alteration in muscarinic receptor density following acute DFP treatment was not accompanied by a change in a particular muscarinic receptor binding conformation. Furthermore, the magnitude of muscarinic receptor-mediated phosphoinositide hydrolysis was unchanged following short-term DFP treatment suggesting that a physiological desensitization in this response does not accompany acute down-regulation of [3H]NMS binding sites.


Subject(s)
Isoflurophate/pharmacology , Phosphatidylinositols/metabolism , Receptors, Muscarinic/drug effects , Animals , Cerebral Cortex/drug effects , Hydrolysis , In Vitro Techniques , Male , Mice , Mice, Inbred ICR , N-Methylscopolamine , Parasympatholytics/pharmacology , Pirenzepine/metabolism , Scopolamine Derivatives/pharmacology
18.
Eur J Pharmacol ; 132(2-3): 147-54, 1986 Dec 16.
Article in English | MEDLINE | ID: mdl-3816972

ABSTRACT

The effect of the irreversible acetylcholinesterase inhibitor diisopropylfluorophosphate (DFP) on mouse brain muscarinic acetylcholine receptors was assessed using the muscarinic antagonists [3H]N-methylscopolamine [( 3H]NMS) and [3H]quinuclidinyl benzilate [( 3H]QNB). No alteration in the maximal binding capacity (Bmax) or equilibrium dissociation constant (KD) was observed in brain homogenates obtained from mice 12 h after a single injection of DFP when [3H]NMS was employed as the ligand. However, one administration of DFP produced a 24 and 33% decrease in Bmax as measured by [3H]NMS binding after 18 and 24 h, respectively. A similar decrease in Bmax was found after two (31%) and three (29%) days of daily DFP treatment. On the other hand, Scatchard analysis using [3H]QNB binding in the brain revealed no difference in KD or Bmax between untreated and 24 h DFP-treated mice. Thus, there is a differential alteration in mouse brain muscarinic acetylcholine receptors using these two ligands which suggests that [3H]NMS binding sites are more sensitive to regulation following acute organophosphate administration.


Subject(s)
Brain Chemistry/drug effects , Organophosphorus Compounds/pharmacology , Receptors, Muscarinic/drug effects , Scopolamine Derivatives/pharmacology , Animals , Cell Membrane/metabolism , In Vitro Techniques , Isoflurophate/pharmacology , Kinetics , Male , Mice , Mice, Inbred ICR , N-Methylscopolamine , Quinuclidinyl Benzilate
19.
Eur J Pharmacol ; 131(2-3): 237-47, 1986 Nov 19.
Article in English | MEDLINE | ID: mdl-3816954

ABSTRACT

In the present work, we studied the details of the interaction of the nonclassical muscarinic receptor antagonist pirenzepine with [3H]quinuclidinyl benzilate binding sites in rat brain homogenates. Pirenzepine showed biphasic competition curves with a Hill coefficient lower than unity, and these curves were better described according to a two-site receptor model. The affinities and the relative preponderance of these sites were constant at different ligand concentrations, in accordance with a competitive type of interaction. Similarly, pirenzepine did not influence the rate of dissociation of the [3H]quinuclidinyl benzilate-receptor complex, even at relatively high concentrations. However, although low concentrations of pirenzepine decreased the affinity of [3H]quinuclidinyl benzilate for the receptor without affecting the density of the binding sites, higher concentrations of the antagonist decreased the receptor number in a reversible fashion. Schild plots of these data indicated an apparent deviation from simple competition in this experimental design, an observation which can be attributed to the selectivity of pirenzepine for different receptor subtypes. Furthermore, pirenzepine, at concentrations high enough to saturate both its high- and low-affinity sites protected [3H]quinuclidinyl benzilate binding sites in the brain against irreversible alkylation by propylbenzilylcholine mustard. Therefore, our data support a competitive nature of interaction of pirenzepine with rat brain muscarinic receptors.


Subject(s)
Brain Chemistry/drug effects , Pirenzepine/pharmacology , Receptors, Cholinergic/drug effects , Receptors, Muscarinic/drug effects , Animals , Binding, Competitive/drug effects , In Vitro Techniques , Kinetics , Male , Quinuclidinyl Benzilate/pharmacology , Rats , Rats, Inbred Strains
20.
J Pharmacol Exp Ther ; 238(3): 916-23, 1986 Sep.
Article in English | MEDLINE | ID: mdl-3018226

ABSTRACT

The effects of brief incubation with carbamylcholine on subsequent binding of [3H]N-methylscopolamine were investigated in mouse neuroblastoma cells (clone N1E-115). This treatment demonstrated that the muscarinic receptors in this neuronal clone can be divided into two types; one which is readily susceptible to regulation by receptor agonists, whereas the other is resistant in this regard. In control cells, both pirenzepine and carbamylcholine interacted with high- and low-affinity subsets of muscarinic receptors. Computer-assisted analysis of the competition between pirenzepine and carbamylcholine with [3H]N-methylscopolamine showed that the receptor sites remaining upon desensitization are composed mainly of pirenzepine low-affinity and agonist high-affinity binding sites. Furthermore, there was an excellent correlation between the ability of various muscarinic receptor agonists to induce a decrease in consequent [3H]N-methylscopolamine binding and their efficacy in stimulating cyclic GMP synthesis in these cells. Thus, only the agonists that are known to recognize the receptor's low-affinity conformation in order to elicit increases in cyclic GMP levels were capable of diminishing ligand binding. Taken together, our present results suggest that the receptor population that is sensitive to regulation by agonists includes both the pirenzepine high-affinity and the agonist low-affinity receptor binding states. In addition, the sensitivity of these receptor subsets to rapid regulation by agonists further implicates their involvement in desensitization of muscarinic receptor-mediated cyclic GMP formation.


Subject(s)
Benzodiazepinones/metabolism , Receptors, Muscarinic/analysis , Carbachol/pharmacology , Cells, Cultured , Cyclic GMP/biosynthesis , N-Methylscopolamine , Neuroblastoma/metabolism , Pirenzepine , Protein Conformation , Receptors, Muscarinic/drug effects , Receptors, Muscarinic/metabolism , Scopolamine Derivatives/metabolism , Tritium
SELECTION OF CITATIONS
SEARCH DETAIL
...