Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 276(27): 25427-37, 2001 Jul 06.
Article in English | MEDLINE | ID: mdl-11306580

ABSTRACT

Understanding the molecular mechanisms of agonist-induced trafficking of G-protein-coupled receptors is important because of the essential role of trafficking in signal transduction. We examined the role of the GTPases dynamin 1 and Rab5a in substance P (SP)-induced trafficking and signaling of the neurokinin 1 receptor (NK1R), an important mediator of pain, depression, and inflammation, by studying transfected cells and enteric neurons that naturally express the NK1R. In unstimulated cells, the NK1R colocalized with dynamin at the plasma membrane, and Rab5a was detected in endosomes. SP induced translocation of the receptor into endosomes containing Rab5a immediately beneath the plasma membrane and then in a perinuclear location. Expression of the dominant negative mutants dynamin 1 K44E and Rab5aS34N inhibited endocytosis of SP by 45 and 32%, respectively. Dynamin K44E caused membrane retention of the NK1R, whereas Rab5aS34N also impeded the translocation of the receptor from superficially located to perinuclear endosomes. Both dynamin K44E and Rab5aS34N strongly inhibited resensitization of SP-induced Ca(2+) mobilization by 60 and 85%, respectively, but had no effect on NK1R desensitization. Dynamin K44E but not Rab5aS34N markedly reduced SP-induced phosphorylation of extracellular signal regulated kinases 1 and 2. Thus, dynamin mediates the formation of endosomes containing the NK1R, and Rab5a mediates both endosomal formation and their translocation from a superficial to a perinuclear location. Dynamin and Rab5a-dependent trafficking is essential for NK1R resensitization but is not necessary for desensitization of signaling. Dynamin-dependent but not Rab5a-dependent trafficking is required for coupling of the NK1R to the mitogen-activated protein kinase cascade. These processes may regulate the nociceptive, depressive, and proinflammatory effects of SP.


Subject(s)
GTP Phosphohydrolases/metabolism , Receptors, Neurokinin-1/metabolism , Signal Transduction , rab5 GTP-Binding Proteins/metabolism , Animals , Calcium/metabolism , Dogs , Dynamin I , Dynamins , Endocytosis , Fluorescent Antibody Technique , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Rats , Transfection , Tumor Cells, Cultured
2.
Am J Physiol Cell Physiol ; 280(5): C1097-106, 2001 May.
Article in English | MEDLINE | ID: mdl-11287322

ABSTRACT

An understanding of the mechanisms that regulate signaling by the substance P (SP) or neurokinin 1 receptor (NK1-R) is of interest because of their role in inflammation and pain. By using activators and inhibitors of protein kinase C (PKC) and NK1-R mutations of potential PKC phosphorylation sites, we determined the role of PKC in desensitization of responses to SP. Activation of PKC abolished SP-induced Ca(2+) mobilization in cells that express wild-type NK1-R. This did not occur in cells expressing a COOH-terminally truncated NK1-R (NK1-Rdelta324), which may correspond to a naturally occurring variant, or a point mutant lacking eight potential PKC phosphorylation sites within the COOH tail (NK1-R Ser-338, Thr-339, Ser-352, Ser-387, Ser-388, Ser-390, Ser-392, Ser-394/Ala, NK1-RKC4). Compared with wild-type NK1-R, the t(1/2) of SP-induced Ca(2+) mobilization was seven- and twofold greater in cells expressing NK1-Rdelta324 and NK1-RKC4, respectively. In cells expressing wild-type NK1-R, inhibition of PKC caused a 35% increase in the t(1/2) of SP-induced Ca(2+) mobilization. Neither inhibition of PKC nor receptor mutation affected desensitization of Ca(2+) mobilization to repeated challenge with SP or SP-induced endocytosis of the NK1-R. Thus PKC regulates SP-induced Ca(2+) mobilization by full-length NK1-R and does not regulate a naturally occurring truncated variant. PKC does not mediate desensitization to repeated stimulation or endocytosis of the NK1-R.


Subject(s)
Calcium Signaling/physiology , Protein Kinase C/metabolism , Receptors, Neurokinin-1/physiology , Substance P/pharmacology , Amino Acid Sequence , Amino Acid Substitution , Animals , Base Sequence , Calcium Signaling/drug effects , Cell Line , DNA Primers , Enzyme Activation , Enzyme Inhibitors/pharmacology , Indoles , Kidney , Maleimides , Molecular Sequence Data , Mutagenesis, Site-Directed , Phorbol 12,13-Dibutyrate/pharmacology , Phosphorylation , Phosphoserine/metabolism , Phosphothreonine/metabolism , Protein Conformation , Rats , Receptors, Neurokinin-1/chemistry , Receptors, Neurokinin-1/drug effects , Recombinant Proteins/chemistry , Recombinant Proteins/drug effects , Recombinant Proteins/metabolism , Sequence Deletion , Transfection
3.
J Neuroendocrinol ; 13(3): 279-87, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11207943

ABSTRACT

Agonist-induced endocytosis of somatostatin receptors determines subsequent cellular responsiveness to peptide agonist and influences somatostatin receptor scintigraphy, a technique to image various tumours. We examined the internalization of sst3HSV, an epitope-tagged type 3 somatostatin receptor, in transfected rat neuroendocrine insulinoma cells. Stimulation of these cells with somatostatin induced trafficking of coexpressed enhanced green fluorescence protein/beta-arrestin1 fusion protein and sst3HSV to colocalize in the same endocytic vesicles. Coexpression of a dominant negative mutant of the arrestin fusion protein with the receptor blocked the internalization of sst3HSV. Stimulation with somatostatin also induced the transient translocation of alpha-adaptin, a component of the adaptor protein complex 2, to the plasma membrane. alpha-adaptin and clathrin colocalized with the receptor. By electron microscopy, we observed internalized sst3 in clathrin coated pits, endosomes and at the limiting membrane of multivesicular bodies, a location typical for receptors being recycled. Concordantly, we observed sst3HSV colocalized with Rab11 in a perinuclear compartment which is likely to correspond to the pericentriolar recycling endosome. Thus, agonist-induced endocytosis of sst3 depends on its interaction with beta-arrestin, involves the adaptor protein complex 2 and proceeds via clathrin coated vesicles to the recycling compartment.


Subject(s)
Arrestins/physiology , Clathrin-Coated Vesicles/physiology , Endocytosis/drug effects , Receptors, Somatostatin/metabolism , Adaptor Protein Complex alpha Subunits , Adaptor Proteins, Vesicular Transport , Animals , Arrestins/genetics , Endosomes/chemistry , Endosomes/metabolism , Fluorescent Antibody Technique , Green Fluorescent Proteins , Immunohistochemistry , Insulinoma , Luminescent Proteins/genetics , Membrane Proteins/metabolism , Microscopy, Electron , Microtubules/metabolism , Microtubules/ultrastructure , Mutation , Pancreatic Neoplasms , Rats , Receptors, Somatostatin/analysis , Recombinant Fusion Proteins/metabolism , Somatostatin/pharmacology , Transfection , Tumor Cells, Cultured , beta-Arrestins , rab GTP-Binding Proteins/analysis
4.
Proc Natl Acad Sci U S A ; 97(20): 11086-91, 2000 Sep 26.
Article in English | MEDLINE | ID: mdl-10995467

ABSTRACT

A requirement for scaffolding complexes containing internalized G protein-coupled receptors and beta-arrestins in the activation and subcellular localization of extracellular signal-regulated kinases 1 and 2 (ERK1/2) has recently been proposed. However, the composition of these complexes and the importance of this requirement for function of ERK1/2 appear to differ between receptors. Here we report that substance P (SP) activation of neurokinin-1 receptor (NK1R) stimulates the formation of a scaffolding complex comprising internalized receptor, beta-arrestin, src, and ERK1/2 (detected by gel filtration, immunoprecipitation, and immunofluorescence). Inhibition of complex formation, by expression of dominant-negative beta-arrestin or a truncated NK1R that fails to interact with beta-arrestin, inhibits both SP-stimulated endocytosis of the NK1R and activation of ERK1/2, which is required for the proliferative and antiapoptotic effects of SP. Thus, formation of a beta-arrestin-containing complex facilitates the proliferative and antiapoptotic effects of SP, and these effects of SP could be diminished in cells expressing truncated NK1R corresponding to a naturally occurring variant.


Subject(s)
Apoptosis/drug effects , Apoptosis/physiology , Arrestins/physiology , Receptors, Neurokinin-1/physiology , Signal Transduction/drug effects , Substance P/pharmacology , Animals , Cell Division/drug effects , Cell Division/physiology , Cell Line , MAP Kinase Signaling System , Rats , beta-Arrestins
5.
Biochem Soc Trans ; 28(4): 419-26, 2000.
Article in English | MEDLINE | ID: mdl-10961932

ABSTRACT

Biological responses to neuropeptides are rapidly attenuated by overlapping mechanisms that include peptide degradation by cell-surface proteases, receptor uncoupling from heterotrimeric G-proteins and receptor endocytosis. We have investigated the mechanisms that terminate the proinflammatory effects of the neuropeptide substance P (SP), which are mediated by the neurokinin 1 receptor (NK1R). Neutral endopeptidase degrades SP in the extracellular fluid and is one of the first mechanisms to terminate signalling. G-protein receptor kinases and second-messenger kinases phosphorylate the NK1R to permit interaction with beta-arrestins, which uncouple the receptor from G-proteins to terminate the signal. SP-induces NK1R endocytosis by a beta-arrestin-dependent mechanism, which also involves the GTPases dynamin and Rab5a. Endocytosis contributes to desensitization by depleting receptors from the cell surface. Disruption of these mechanisms results in uncontrolled stimulation and disease. Thus the deletion of neutral endopeptidase in mice exacerbates inflammation of many tissues. There are similarities and distinct differences in the mechanisms that regulate signalling by neuropeptide receptors and other G-protein-coupled receptors, in particular those that are activated irreversibly by proteolysis.


Subject(s)
Endopeptidases/metabolism , Receptors, Neuropeptide/metabolism , Signal Transduction , Substance P/metabolism , Animals , Arrestins/metabolism , Cell Membrane/metabolism , Dynamins , Endocytosis , GTP Phosphohydrolases/metabolism , Humans , Mast Cells/metabolism , Mice , Models, Biological , Neprilysin/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Receptor, PAR-2 , Receptors, Neurokinin-1/metabolism , Receptors, Thrombin/metabolism , Serine Endopeptidases/metabolism , Time Factors , Tryptases , beta-Arrestins , rab5 GTP-Binding Proteins/metabolism
6.
J Cell Biol ; 148(6): 1267-81, 2000 Mar 20.
Article in English | MEDLINE | ID: mdl-10725339

ABSTRACT

Recently, a requirement for beta-arrestin-mediated endocytosis in the activation of extracellular signal-regulated kinases 1 and 2 (ERK1/2) by several G protein-coupled receptors (GPCRs) has been proposed. However, the importance of this requirement for function of ERK1/2 is unknown. We report that agonists of Galphaq-coupled proteinase-activated receptor 2 (PAR2) stimulate formation of a multiprotein signaling complex, as detected by gel filtration, immunoprecipitation and immunofluorescence. The complex, which contains internalized receptor, beta-arrestin, raf-1, and activated ERK, is required for ERK1/2 activation. However, ERK1/2 activity is retained in the cytosol and neither translocates to the nucleus nor causes proliferation. In contrast, a mutant PAR2 (PAR2deltaST363/6A), which is unable to interact with beta-arrestin and, thus, does not desensitize or internalize, activates ERK1/2 by a distinct pathway, and fails to promote both complex formation and cytosolic retention of the activated ERK1/2. Whereas wild-type PAR2 activates ERK1/2 by a PKC-dependent and probably a ras-independent pathway, PAR2(deltaST363/6A) appears to activate ERK1/2 by a ras-dependent pathway, resulting in increased cell proliferation. Thus, formation of a signaling complex comprising PAR2, beta-arrestin, raf-1, and activated ERK1/2 might ensure appropriate subcellular localization of PAR2-mediated ERK activity, and thereby determine the mitogenic potential of receptor agonists.


Subject(s)
Arrestins/physiology , Endocytosis , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinases/metabolism , Receptors, Thrombin/physiology , Animals , Calcium/metabolism , Cell Division , Cell Line , Cell Nucleus/physiology , Cell Nucleus/ultrastructure , Cytosol/physiology , Cytosol/ultrastructure , Enzyme Activation , Humans , Kinetics , Microscopy, Confocal , Mitogen-Activated Protein Kinase 3 , Models, Biological , Mutagenesis , Rats , Receptor, PAR-2 , Receptors, Thrombin/genetics , Recombinant Proteins/metabolism , Transfection , beta-Arrestins
7.
J Physiol ; 517 ( Pt 3): 741-56, 1999 Jun 15.
Article in English | MEDLINE | ID: mdl-10358115

ABSTRACT

1. Proteases regulate cells by cleaving proteinase-activated receptors (PARs). Thrombin and trypsin cleave PAR-1 and PAR-2 on neurons and astrocytes of the brain to regulate morphology, growth and survival. We hypothesized that thrombin and mast cell tryptase, which are generated and released during trauma and inflammation, regulate enteric neurons by cleaving PAR-1 and PAR-2. 2. We detected immunoreactive PAR-1 and PAR-2 in > 60 % of neurons from the myenteric plexus of guinea-pig small intestine in primary culture. A large proportion of neurons that expressed substance P, vasoactive intestinal peptide or nitric oxide synthase also expressed PAR-1 and PAR-2. We confirmed expression of PAR-1 and PAR-2 in the myenteric plexus by RT-PCR using primers based on sequences of cloned guinea-pig receptors. 3. Thrombin, trypsin, tryptase, a filtrate from degranulated mast cells, and peptides corresponding to the tethered ligand domains of PAR-1 and PAR-2 increased [Ca2+]i in > 50 % of cultured myenteric neurons. Approximately 60 % of neurons that responded to PAR-1 agonists responded to PAR-2 agonists, and > 90 % of PAR-1 and PAR-2 responsive neurons responded to ATP. 4. These results indicate that a large proportion of myenteric neurons that express excitatory and inhibitory neurotransmitters and purinoceptors also express PAR-1 and PAR-2. Thrombin and tryptase may excite myenteric neurons during trauma and inflammation when prothrombin is activated and mast cells degranulate. This novel action of serine proteases probably contributes to abnormal neurotransmission and motility in the inflamed intestine.


Subject(s)
Myenteric Plexus/physiology , Neurons/physiology , Receptors, Thrombin/physiology , Serine Endopeptidases/metabolism , Thrombin/metabolism , Animals , Animals, Newborn , Benzimidazoles/pharmacology , Calcium/metabolism , Cells, Cultured , Chymases , Cloning, Molecular , Guinea Pigs , Homeostasis , Immunohistochemistry , Intestine, Small/innervation , Male , Neurons/cytology , Neurons/drug effects , Oligopeptides/pharmacology , Receptor, PAR-1 , Receptor, PAR-2 , Receptors, Thrombin/analysis , Receptors, Thrombin/genetics , Recombinant Proteins/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Tryptases , Vasoactive Intestinal Peptide/analysis
8.
J Biol Chem ; 274(26): 18524-35, 1999 Jun 25.
Article in English | MEDLINE | ID: mdl-10373461

ABSTRACT

Proteases cleave proteinase-activated receptors (PARs) to expose N-terminal tethered ligands that bind and activate the cleaved receptors. The tethered ligand, once exposed, is always available to interact with its binding site. Thus, efficient mechanisms must prevent continuous activation, including receptor phosphorylation and uncoupling from G-proteins, receptor endocytosis, and lysosomal degradation. beta-Arrestins mediate uncoupling and endocytosis of certain neurotransmitter receptors, which are activated in a reversible manner. However, the role of beta-arrestins in trafficking of PARs, which are irreversibly activated, and the effects of proteases on the subcellular distribution of beta-arrestins have not been examined. We studied trafficking of PAR2 and beta-arrestin1 coupled to green fluorescent protein. Trypsin induced the following: (a) redistribution of beta-arrestin1 from the cytosol to the plasma membrane, where it co-localized with PAR2; (b) internalization of beta-arrestin1 and PAR2 into the same early endosomes; (c) redistribution of beta-arrestin1 to the cytosol concurrent with PAR2 translocation to lysosomes; and (d) mobilization of PAR2 from the Golgi apparatus to the plasma membrane. Overexpression of a C-terminal fragment of beta-arrestin-319-418, which interacts constitutively with clathrin but does not bind receptors, inhibited agonist-induced endocytosis of PAR2. Our results show that beta-arrestins mediate endocytosis of PAR2 and support a role for beta-arrestins in uncoupling of PARs.


Subject(s)
Arrestins/metabolism , Endocytosis , Receptors, Thrombin/metabolism , Animals , Blotting, Western , Calcium/metabolism , Cell Line , Green Fluorescent Proteins , Indicators and Reagents , Luminescent Proteins , Rabbits , Rats , Receptor, PAR-2 , Trypsin/metabolism , Tumor Cells, Cultured , beta-Arrestin 1 , beta-Arrestins
9.
J Biol Chem ; 274(23): 16257-68, 1999 Jun 04.
Article in English | MEDLINE | ID: mdl-10347182

ABSTRACT

Agonist-induced redistribution of G-protein-coupled receptors (GPCRs) and beta-arrestins determines the subsequent cellular responsiveness to agonists and is important for signal transduction. We examined substance P (SP)-induced trafficking of beta-arrestin1 and the neurokinin-1 receptor (NK1R) in KNRK cells in real time using green fluorescent protein. Green fluorescent protein did not alter function or localization of the NK1R or beta-arrestin1. SP induced (a) striking and rapid (<1 min) translocation of beta-arrestin1 from the cytosol to the plasma membrane, which preceded NK1R endocytosis; (b) redistribution of the NK1R and beta-arrestin1 into the same endosomes containing SP and the transferrin receptor (2-10 min); (c) prolonged colocalization of the NK1R and beta-arrestin1 in endosomes (>60 min); (d) gradual resumption of the steady state distribution of the NK1R at the plasma membrane and beta-arrestin1 in the cytosol (4-6 h). SP stimulated a similar redistribution of immunoreactive beta-arrestin1 and beta-arrestin2. In contrast, SP did not affect Galphaq/11 distribution, which remained at the plasma membrane. Expression of the dominant negative beta-arrestin319-418 inhibited SP-induced endocytosis of the NK1R. Thus, SP induces rapid translocation of beta-arrestins to the plasma membrane, where they participate in NK1R endocytosis. beta-Arrestins colocalize with the NK1R in endosomes until the NK1R recycles and beta-arrestins return to the cytosol.


Subject(s)
Arrestins/metabolism , Endocytosis , Receptors, Neurokinin-1/metabolism , Substance P/metabolism , Animals , Arrestins/genetics , Biological Transport , Cell Line , Green Fluorescent Proteins , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Rabbits , Rats , Receptors, Neurokinin-1/genetics , Recombinant Fusion Proteins/metabolism , Transfection , beta-Arrestins
11.
Am J Physiol ; 274(6): C1429-52, 1998 06.
Article in English | MEDLINE | ID: mdl-9696685

ABSTRACT

Although serine proteases are usually considered to act principally as degradative enzymes, certain proteases are signaling molecules that specifically regulate cells by cleaving and triggering members of a new family of proteinase-activated receptors (PARs). There are three members of this family, PAR-1 and PAR-3, which are receptors for thrombin, and PAR-2, a receptor for trypsin and mast cell tryptase. Proteases cleave within the extracellular NH2-terminus of their receptors to expose a new NH2-terminus. Specific residues within this tethered ligand domain interact with extracellular domains of the cleaved receptor, resulting in activation. In common with many G protein-coupled receptors, PARs couple to multiple G proteins and thereby activate many parallel mechanisms of signal transduction. PARs are expressed in multiple tissues by a wide variety of cells, where they are involved in several pathophysiological processes, including growth and development, mitogenesis, and inflammation. Because the cleaved receptor is physically coupled to its agonist, efficient mechanisms exist to terminate signaling and prevent uncontrolled stimulation. These include cleavage of the tethered ligand, receptor phosphorylation and uncoupling from G proteins, and endocytosis and lysosomal degradation of activated receptors.


Subject(s)
DNA-Binding Proteins/physiology , Receptors, Cell Surface/physiology , Receptors, Thrombin/physiology , Saccharomyces cerevisiae Proteins , Serine Endopeptidases/physiology , Signal Transduction , Transcription Factors/physiology , Amino Acid Sequence , Animals , GTP-Binding Proteins/metabolism , Humans , Molecular Sequence Data , Protein Binding/physiology , Receptor, PAR-2 , Receptors, Cell Surface/agonists , Receptors, Thrombin/agonists
12.
J Clin Invest ; 100(6): 1383-93, 1997 Sep 15.
Article in English | MEDLINE | ID: mdl-9294103

ABSTRACT

Proteinase-activated receptor-2 (PAR-2) is a G protein-coupled receptor that is cleaved and activated by trypsin-like enzymes. PAR-2 is highly expressed by small intestinal enterocytes where it is activated by luminal trypsin. The location, mechanism of activation, and biological functions of PAR-2 in the colon, however, are unknown. We localized PAR-2 to the muscularis externa of the rat colon by immunofluorescence. Myocytes in primary culture also expressed PAR-2, assessed by immunofluorescence and RT-PCR. Trypsin, SLIGRL-NH2 (corresponding to the PAR-2 tethered ligand), mast cell tryptase, and a filtrate of degranulated mast cells stimulated a prompt increase in [Ca2+]i in myocytes. The response to tryptase and the mast cell filtrate was inhibited by the tryptase inhibitor BABIM, and abolished by desensitization of PAR-2 with trypsin. PAR-2 activation inhibited the amplitude of rhythmic contractions of strips of rat colon. This response was unaffected by indomethacin, l-NG-nitroarginine methyl ester, a bradykinin B2 receptor antagonist and tetrodotoxin. Thus, PAR-2 is highly expressed by colonic myocytes where it may be cleaved and activated by mast cell tryptase. This may contribute to motility disturbances of the colon during conditions associated with mast cell degranulation.


Subject(s)
Mast Cells/enzymology , Receptors, Cell Surface/physiology , Serine Endopeptidases/metabolism , Adrenergic beta-Antagonists/pharmacology , Animals , Benzimidazoles/pharmacology , Bradykinin/analogs & derivatives , Bradykinin/pharmacology , Calcium/metabolism , Cells, Cultured , Chymases , Colon/chemistry , Cyclooxygenase Inhibitors/pharmacology , Dinoprostone/pharmacology , Dose-Response Relationship, Drug , Fluorescent Antibody Technique, Indirect , Gastrointestinal Motility/drug effects , In Vitro Techniques , Indomethacin/pharmacology , Inflammation Mediators/pharmacology , Leupeptins/pharmacology , Muscle, Smooth/chemistry , NG-Nitroarginine Methyl Ester/pharmacology , Nitroprusside/pharmacology , Rats , Receptor, PAR-2 , Receptors, Cell Surface/analysis , Serine Endopeptidases/pharmacology , Substance P/pharmacology , Tetrodotoxin/pharmacology , Time Factors , Trypsin/pharmacology , Tryptases
13.
Mol Immunol ; 34(12-13): 829-38, 1997.
Article in English | MEDLINE | ID: mdl-9464518

ABSTRACT

In a previous study (Boquet et al., 1995, Molec. Immunol. 32, 303-308) we observed remarkable inversions of hydropathic profiles between complementarity determining regions (CDRs) of an anti-substance P monoclonal antibody (SP31) and the corresponding 5 amino acid C-terminal peptide epitope. Here we demonstrate the importance this hydropathic complementarity by measuring the immunoreactivity of SP-related peptides which have been modified in their C-terminal parts so that hydropathic profile has been conserved (by substituting amino acids in the epitope) or modified (by mixing the sequence of amino acids in the epitope). Experiments performed in equilibrium conditions using a competitive enzyme immunoassay showed that most of the peptides conserving the hydropathic profile of SP epitope were recognized by mAb SP31 (even if marked variations in affinity were observed), while those for which the hydropathic profile was modified exhibited very low or undetectable affinity. The kinetic parameters (ka and kd) of peptide-antibody interactions were determined using Surface Plasmon Resonance technology (BIACORE 2000). These measurements showed that all peptides recognized by mAb SP31 had similar association rate constants (close to 2 x 10[5] M[-1] s[-1]), differences in binding affinities essentially resulting from differences in dissociation rate constants (ranging from 1.61 x 10[-4] to 1.15 x 10[-1] s[-1]). From these results, it was concluded that hydropathic complementarity between the epitope and the paratope could be a necessary but not sufficient condition for establishing high-affinity binding. We hypothesize that hydropathic interactions may play a critical role during the first contacts between antibody CDRs and the peptide, possibly by favouring reorganization of water molecules at the antibody-peptide interface.


Subject(s)
Antibodies, Monoclonal/immunology , Substance P/immunology , Amino Acid Sequence , Antibody Specificity , Antigen-Antibody Reactions , Biosensing Techniques , Cross Reactions , Enzyme-Linked Immunosorbent Assay , Epitope Mapping , Kinetics , Peptide Fragments/immunology , Structure-Activity Relationship , Substance P/chemistry
14.
J Neuroimmunol ; 76(1-2): 1-9, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9184627

ABSTRACT

Monoclonal antibodies to the binding site of the NK1 receptor for the neuropeptide substance P were produced in mice using the complementary or antisense peptide methodology. Among several anti-peptide monoclonal antibodies, we selected the mAb12 antibody which specifically crossreacted, through its paratope, with a binding site present on membranes from rat parotid gland cells, with an affinity close to 2 x 10(-7) M and with membranes from CHO cells expressing human brain NK1 receptors. Immunocytochemical investigations using mAb12 revealed immunostaining whose distribution in the dorsal horns of rat spinal cord fits well with the known location of NK1 receptors. In both biochemical and immunocytochemical experiments, the competition occurring between the antibody and substance P, or a substance P-protein conjugate, indicates that mAb12 recognizes a membrane epitope located at or near the substance P binding domain on the NK1 receptor. Immunization of mice with mAb12 led to the production of specific anti-substance P antibodies, again suggesting that mAb12 shares common structural features with the neuropeptide. This monoclonal antibody can now be used in further biochemical or cytochemical characterizations of NK1 receptors. Owing to its fine specificity, mAb12 could also serve as a molecular model for designing peptides, possibly displaying pharmacological properties in the various processes in which substance P is involved, e.g. immunomodulation, inflammation or chronic pain.


Subject(s)
Antibodies, Monoclonal/immunology , Receptors, Neurokinin-1/analysis , Substance P/immunology , Animals , Binding Sites , CHO Cells , Cricetinae , Female , Humans , Immunization , Immunohistochemistry , Male , Mice , Rats , Rats, Wistar , Receptors, Neurokinin-1/immunology
15.
Biopolymers ; 39(1): 67-74, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8924627

ABSTRACT

The topography of the binding site of a monoclonal anti-substance P antibody directed toward the C-terminal pentapeptide of substance P, Arg-Pro-Lys-Pro-Gln-Gln-Phe-Phe-Gly-Leu-Met-NH2, was analyzed further using a wide range of constrained analogues of substance P. Results obtained in the present study show the following: (a) The binding subsites of Phe7 and Phe8 are large and deep, accommodating various side chains, including nonaromatic amino acids. (b) In contrast, the binding pockets for Gly-Leu-Met-NH2 appear more restrictive. Consequently, five residues in the peptide are necessary for the high binding affinity to the antibody, the C-terminal tripeptide determining the binding specificity. These data, which appear to contradict those previously published, illustrate the limits of conclusions drawn from studies generally carried out using exclusively Ala-substituted peptides. In addition, the present results indicate that the topography of the binding site of this monoclonal antibody differs from that of the specific substance P neurokinin-1 receptor, in agreement with the differences observed in the fine specificities of these two substance P binding macromolecules.


Subject(s)
Antibodies, Monoclonal/chemistry , Binding Sites, Antibody , Substance P/chemistry , Substance P/immunology , Antibodies, Monoclonal/immunology , Binding Sites , Oligopeptides/chemistry , Oligopeptides/immunology , Oligopeptides/metabolism , Receptors, Neurokinin-1/chemistry , Receptors, Neurokinin-1/metabolism , Substance P/analogs & derivatives , Substance P/metabolism
16.
J Chem Neuroanat ; 9(1): 65-77, 1995 Jul.
Article in English | MEDLINE | ID: mdl-8527039

ABSTRACT

A monoclonal antibody directed against a peptide (PS5) specified by RNA complementary to the mRNA coding for substance P (SP), was used to label SP receptors in the rat spinal cord as demonstrated by light and electron microscopy. An immunocytochemical method (avidin-biotin-peroxidase) was used on vibratome sections from rats perfused with paraformaldehyde. Immunoreactivity was observed principally in the two superficial layers of the dorsal horn, in lamina X and the region of motoneurons. The labeling was absent when the antibody was preincubated with the complementary peptide (PS5) used as immunogen. Competition between the anti-complementary peptide antibody and different ligands was tested by preincubation of tissue sections with the ligand in the presence of peptidase inhibitors before addition of the antibody. A specific agonist (SP) or antagonist (spantide, RP 67580) at 10(-6)M led to total absence of labeling. These results indicate that under our experimental conditions, the anti-complementary peptide antibody recognizes a SP binding site in the rat spinal cord. Electron microscopic study of the two superficial laminae of the dorsal horn showed that immunolabeling was mainly localized extracellularly at apposing neuronal plasma membranes. It was mostly associated with axodendritic or axosomatic appositions. Occasionally labeling was observed between two axon terminals. In all cases, these appositions were non-junctional. Generally, neuronal processes involved in these appositions did not contain large granular vesicles. These observations suggest that SP may act in a diffuse, nonsynaptic manner probably on targets distant from SP release sites.


Subject(s)
Receptors, Neurokinin-1/analysis , Spinal Cord/chemistry , Amino Acid Sequence , Animals , Antibodies, Monoclonal , Cell Membrane/chemistry , Cell Membrane/ultrastructure , Immunohistochemistry , Indoles/pharmacology , Isoindoles , Male , Mice , Microscopy, Electron , Molecular Sequence Data , Neurokinin-1 Receptor Antagonists , RNA, Complementary , Rats , Rats, Wistar , Receptors, Neurokinin-1/agonists , Spinal Cord/ultrastructure , Substance P/analogs & derivatives , Substance P/immunology , Substance P/metabolism , Substance P/pharmacology
18.
J Neurochem ; 63(1): 333-43, 1994 Jul.
Article in English | MEDLINE | ID: mdl-7515949

ABSTRACT

We used complementary peptide methodology to obtain antibodies against the receptor for the neuropeptide substance P, specifically directed at the ligand-binding domain. Rabbits were immunized with two distinct peptides derived from the sequence of the RNA complementary to the mRNA for substance P. Binding experiments revealed that antipeptide polyclonal antibodies were able to recognize, through their paratope, a specific binding site on the rat parotid cell membranes. Substance P and antibodies competed for this binding site, because preincubation of membranes in the presence of substance P significantly reduced antibody binding, and conversely, preincubation of membranes in the presence of antibodies partly inhibited the binding of radioiodinated substance P. Immunocytochemical experiments performed on the rat cervical spinal cord show that the distribution of labeling by antibodies is similar to that observed by conventional autoradiography using 125I-substance P. Here again, control experiments demonstrated that antibodies and substance P were competing for the same binding site on the spinal cord. These biochemical and immunocytochemical data indicate that antipeptide antibodies recognize a substance P membrane binding site in nervous and nonnervous mammalian tissues. This site is likely to correspond to the NK1 specific receptor for substance P.


Subject(s)
Antibodies/immunology , Parotid Gland/chemistry , Receptors, Neurokinin-1/analysis , Spinal Cord/chemistry , Substance P/analysis , Substance P/immunology , Amino Acid Sequence , Animals , Antibodies/analysis , Autoradiography , Base Sequence , Binding Sites , Immunohistochemistry , Iodine Radioisotopes , Ligands , Molecular Sequence Data , Parotid Gland/metabolism , Parotid Gland/ultrastructure , Rats , Rats, Wistar , Receptors, Neurokinin-1/metabolism , Spinal Cord/metabolism , Spinal Cord/ultrastructure , Substance P/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...