Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Int J Mol Sci ; 22(8)2021 Apr 08.
Article in English | MEDLINE | ID: mdl-33917689

ABSTRACT

The value of bone marrow aspirate concentrates for treatment of human knee cartilage lesions is unclear. Most of the studies were performed with intra-articular injections. However, subchondral bone plays an important role in the progression of osteoarthritis. We investigated by a literature review whether joint, subchondral bone, or/and scaffolds implantation of fresh autologous bone marrow aspirate concentrated (BMAC) containing mesenchymal stem cells (MSCs) would improve osteoarthritis (OA). There is in vivo evidence that suggests that all these different approaches (intra-articular injections, subchondral implantation, scaffolds loaded with BMAC) can improve the patient. This review analyzes the evidence for each different approach to treat OA. We found that the use of intra-articular injections resulted in a significant relief of pain symptoms in the short term and was maintained in 12 months. However, the clinical trials indicate that the application of autologous bone marrow concentrates in combination with scaffolds or in injection in the subchondral bone was superior to intra-articular injection for long-term results. The tendency of MSCs to differentiate into fibrocartilage affecting the outcome was a common issue faced by all the studies when biopsies were performed, except for scaffolds implantation in which some hyaline cartilage was found. The review suggests also that both implantation of subchondral BMAC and scaffolds loaded with BMAC could reduce the need for further surgery.


Subject(s)
Bone Marrow Transplantation , Bone Regeneration , Cartilage, Articular/pathology , Osteoarthritis, Knee/pathology , Osteoarthritis, Knee/therapy , Bone Marrow Transplantation/methods , Disease Management , Humans , Hyaline Cartilage/pathology , Injections, Intra-Articular , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Osteoarthritis, Knee/etiology , Tissue Engineering , Treatment Outcome
2.
Int Orthop ; 42(7): 1755-1767, 2018 07.
Article in English | MEDLINE | ID: mdl-29882123

ABSTRACT

INTRODUCTION: Human spontaneous osteonecrosis of the knee (SPONK) is still challenging as the current treatments do not allow the production of hyaline cartilage tissue. The aim of the present study was to explore the therapeutic potential of cartilage regeneration using a new biphasic scaffold (type I collagen/hydroxyapatite) previously loaded or not with concentrated bone marrow cells. MATERIAL AND METHODS: Female rabbits were operated of one knee to create articular lesions of the trochlea (three holes of 4 × 4mm). The holes were left empty in the control group or were filled with the scaffold alone or the scaffold previously loaded with concentrated bone marrow cells. After two months, rabbits were sacrificed and the structure of the newly formed tissues were evaluated by macroscopic, MRI, and immunohistochemistry analyses. RESULTS: Macroscopic and MRI evaluation of the knees did not show differences between the three groups (p > 0.05). However, histological analysis demonstrated that a higher O'Driscoll score was obtained in the two groups treated with the scaffold, as compared to the control group (p < 0.05). The number of cells in treated area was higher in scaffold groups compared to the control group (p < 0.05). There was no difference for intensity of collagen type II between the groups (p > 0.05) but subchondral bone repair was significantly thicker in scaffold-treated groups than in the control group (1 mm for the control group vs 2.1 and 2.6 mm for scaffold groups). Furthermore, we observed that scaffolds previously loaded with concentrated bone marrow were more reabsorbed (p < 0.05). CONCLUSION: The use of a biphasic scaffold previously loaded with concentrated bone marrow significantly improves cartilage lesion healing.


Subject(s)
Cartilage, Articular/surgery , Knee Joint/physiopathology , Mesenchymal Stem Cell Transplantation/methods , Tissue Scaffolds , Animals , Bone Regeneration/physiology , Cartilage, Articular/metabolism , Cartilage, Articular/physiopathology , Collagen Type I/pharmacology , Collagen Type II/metabolism , Durapatite/pharmacology , Female , Immunohistochemistry , Knee Joint/metabolism , Knee Joint/surgery , Magnetic Resonance Imaging , Rabbits
3.
J Biol Chem ; 285(40): 30951-8, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20647307

ABSTRACT

The glucagon receptor belongs to the B family of G-protein coupled receptors. Little structural information is available about this receptor and its association with glucagon. We used the substituted cysteine accessibility method and three-dimensional molecular modeling based on the gastrointestinal insulinotropic peptide and glucagon-like peptide 1 receptor structures to study the N-terminal domain of this receptor, a central element for ligand binding and specificity. Our results showed that Asp(63), Arg(116), and Lys(98) are essential for the receptor structure and/or ligand binding because mutations of these three residues completely disrupted or markedly impaired the receptor function. In agreement with these data, our models revealed that Asp(63) and Arg(116) form a salt bridge, whereas Lys(98) is engaged in cation-π interactions with the conserved tryptophans 68 and 106. The native receptor could not be labeled by hydrophilic cysteine biotinylation reagents, but treatment of intact cells with [2-(trimethylammonium)ethyl]methanethiosulfonate increased the glucagon binding site density. This result suggested that an unidentified protein with at least one free cysteine associated with the receptor prevented glucagon recognition and that [2-(trimethylammonium)ethyl]methanethiosulfonate treatment relieved this inhibition. The substituted cysteine accessibility method was also performed on 15 residues selected using the three-dimensional models. Several receptor mutants, despite a relatively high predicted cysteine accessibility, could not be labeled by specific reagents. The three-dimensional models show that these mutated residues are located on one face of the protein. This could be part of the interface between the receptor and the unidentified inhibitory protein, making these residues inaccessible to biotinylation compounds.


Subject(s)
Amino Acid Substitution , Models, Molecular , Receptors, Glucagon/chemistry , Receptors, Glucagon/metabolism , Cell Line , Cysteine/chemistry , Cysteine/genetics , Cysteine/metabolism , Humans , Mutagenesis , Mutation, Missense , Protein Structure, Tertiary , Receptors, Glucagon/genetics
4.
Article in English | MEDLINE | ID: mdl-31708874

ABSTRACT

In bone diseases such as osteonecrosis and osteoporosis, a shift toward a preferential differentiation of mesenchymal stromal cells (MSC) into adipocytes at the expense of the osteoblastic lineage is described, leading to excessive accumulation of adipocytes in the bone marrow of the patients. The influence of cytokines and adipokines secreted by adipocytes on skeletal health is already well-documented but the impact of free fatty acids release on bone cell biology and viability is an emerging concept. We have previously demonstrated that the saturated fatty acid (SFA) palmitate (Palm) is cytotoxic for human MSC (hMSC) and osteoblasts whereas oleate (Ole), a monounsaturated fatty acid (MUFA), has no toxic effect. Moreover, Ole protects cells against lipotoxicity. Our observations led us to propose that the toxicity of the SFA is not correlated to its intracellular accumulation but could rather be related to the intracellular SFA/MUFA ratio, which finally determines the toxic effect of SFA. Therefore, in the present study, we have investigated the potential protective role of the enzyme stearoyl-CoA 9-desaturase 1 (SCD1) against the deleterious effects of Palm. SCD1 is an enzyme responsible for desaturation of SFA to MUFA; its activation could therefore lead to modifications of the intracellular SFA/MUFA ratio. In the present study, we showed that hMSC express SCD1 and liver X receptors (LXRs), transcription factors regulating SCD1 expression. Human MSC treatment with a LXRs agonist triggered SCD1 expression and drastically reduced Palm-induced cell mortality, caspases 3/7 activation, endoplasmic reticulum stress and inflammation. We also observed that, in the presence of Palm, the LXRs agonist provoked lipid droplets formation, augmented the total cellular neutral lipid content but decreased the SFA/MUFA ratio when compared to Palm treatment alone. Addition of an inhibitor of SCD1 activity abrogated the positive effects of the LXRs agonist, suggesting that SCD1 could play a key role in protecting hMSC against lipotoxicity.

5.
Endocrinology ; 158(3): 490-502, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28359085

ABSTRACT

Osteonecrosis of the femoral head (ON) is a multifactorial bone disease that can evolve to a progressive destruction of the hip joint. Different pathogenic processes have been proposed, among them, an increase of bone marrow (BM) fat resulting from adipocyte accumulation. Marrow adipocytes are active BM residents that influence the microenvironment by releasing cytokines, adipokines, and free fatty acids (FA). We explored the impact of palmitate (Palm) and oleate on function and survival of BM-derived mesenchymal stromal cells (MSC) of osteonecrotic patients (ONMSC) and healthy volunteers. Moreover, we analyzed the FA profile of the serum and the BM supernatant fluid (BMSF). We demonstrated that exposure to the saturated FA Palm favored MSC differentiation through the adipogenic lineage at the expense of the osteoblastic phenotype. Moreover, adipogenesis was intensified in ONMSC. The susceptibility to Palm toxicity was aggravated in ONMSC concomitantly with a greater activation of the proapoptotic extracellular signal-regulated kinase pathway. Moreover, cellular mechanisms implicated in the protection against lipotoxicity, such as stearoyl-coenzyme A desaturase 1 and carnitine palmitoyl transferase 1 expression, were dysregulated in ONMSC. Palm-induced interleukin (IL)-6 and IL-8 secretion was also exacerbated in ONMSC. Our results established that, in the serum, the FA profiles were comparable in ON and healthy subjects. However, both the concentrations and the FA composition were modified in the BMSF of ON patients, highlighting a drastic change of the BM microenvironment in ON patients. Altogether, our work suggests that marrow adipocyte enlargement could affect the process of bone remodeling and, therefore, play a role in the pathogenesis of ON.


Subject(s)
Bone Marrow/metabolism , Femur Head Necrosis/blood , Mesenchymal Stem Cells/drug effects , Oleic Acid/toxicity , Palmitic Acid/toxicity , Adipogenesis/drug effects , Adult , Case-Control Studies , Female , Humans , MAP Kinase Signaling System , Male , Oleic Acid/blood , Palmitic Acid/blood
6.
Biochem Pharmacol ; 70(6): 879-87, 2005 Sep 15.
Article in English | MEDLINE | ID: mdl-16076462

ABSTRACT

UNLABELLED: The motilin receptor (MTLR) represents a clinically useful pharmacological target, as agonists binding to the MTLR have gastroprokinetic properties. In order to compare the molecular basis for interaction of the MTLR with motilin and with the non-peptide motilin agonist, erythromycin-A (EM-A), the negatively charged E119 located in the third transmembrane (TM3) region was mutated to D (E119D) and Q (E119Q), respectively, and changes in activity of the mutant receptors were verified. METHODS: Each mutant receptor was stably transfected in CHO-cells containing the Ca2+ indicator apo-aequorin. Receptor activation in response to motilin, EM-A and their analogues was assessed by Ca2+-luminescense. RESULTS: In the E119Q mutant, the Ca2+ response to motilin and EM-A was abolished while in the E119D mutant it was reduced with 62% (motilin) and 81% (EM-A). The pEC50 values were shifted from 9.65+/-0.03 to 7.41+/-0.09 (motilin) and from 6.63+/-0.12 to 4.60+/-0.07 (EM-A). Acetylation of the N-terminal amine group as in [N-acetyl-Phe]1 mot (1-14), decreased the potency 6.3-fold (WT-MTLR) and 148-fold (E119D). Acetylation of EM-A enol ether induced a more pronounced shift in potency: 7943-fold (WT-MTLR) and 1413-fold (E119D). CONCLUSION: The comparable loss of affinity of the mutant receptors for motilin and EM-A indicate that these agonists both interact with the TM3 domain of the MTLR. The results with acetylated derivatives support an ionic interaction between E119 of the MTLR with the N+ of the desosamine sugar in EM-A, but not with the N+ of the free amine group in motilin.


Subject(s)
Erythromycin/metabolism , Motilin/metabolism , Receptors, Gastrointestinal Hormone/metabolism , Receptors, Neuropeptide/metabolism , Amino Acid Sequence , Animals , Binding Sites , Blotting, Western , CHO Cells , Cricetinae , Immunohistochemistry , Molecular Sequence Data , Receptors, Gastrointestinal Hormone/chemistry , Receptors, Neuropeptide/chemistry , Swine
7.
Eur J Pharmacol ; 520(1-3): 70-6, 2005 Sep 27.
Article in English | MEDLINE | ID: mdl-16125696

ABSTRACT

The novel antiepileptic drug levetiracetam ((2S-(2-oxo-1-pyrrolidinyl)butanamide, KEPPRA possesses a specific binding site in brain, which has very recently been identified as the synaptic vesicle protein SV 2 A. The aim of this study was to evaluate the presence of a levetiracetam binding site in the spinal cord and compare its properties to that in rat brain. We used [(3)H]ucb 30889 ((2S)-2-[4-(3-azidophenyl)-2-oxopyrrolidin-1-yl]butanamide), a levetiracetam analogue, to perform binding assays, photoaffinity labelling and autoradiography experiments, and revealed the presence of SV 2 A by Western-blot analysis. [(3)H]ucb 30889 binding kinetics at 4 degrees C were biphasic and saturation binding curves were compatible with the labelling of a homogenous population of binding sites with a K(d) similar to that in brain. Competition curves with ligands known to interact with levetiracetam binding sites and photolabelling experiments indicated that [(3)H]ucb 30889 labels the same 90 kDa protein in both spinal cord and brain. Levetiracetam binding site was localised in the grey matter of the spinal cord and its expression was not modified in a model of neuropathic pain. This study demonstrates the presence of a specific levetiracetam binding site in the rat spinal cord, which is similar to that found in rat brain.


Subject(s)
Anticonvulsants/metabolism , Azides/metabolism , Membrane Glycoproteins/metabolism , Nerve Tissue Proteins/metabolism , Piracetam/analogs & derivatives , Pyrrolidines/metabolism , Spinal Cord/metabolism , Animals , Anticonvulsants/chemistry , Azides/chemistry , Binding Sites , Binding, Competitive , Dose-Response Relationship, Drug , Kinetics , Levetiracetam , Piracetam/chemistry , Piracetam/metabolism , Pyrrolidines/chemistry , Rats , Rats, Sprague-Dawley , Synaptic Vesicles/metabolism
8.
Cell Signal ; 14(8): 689-94, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12020769

ABSTRACT

The vasoactive intestinal peptide receptor VPAC(1) is preferentially coupled to G(alpha s) protein but also increases [Ca(2+)](i) through interaction with G(alpha i)/G(alpha q) protein. We evaluated a panel of full, partial and null agonists for their capability to stimulate adenylate cyclase activity in both intact cells and membrane and [Ca(2+)](i) in intact cells transfected with the reporter gene aequorin. In intact cells, the agonists efficacy for cAMP and calcium increase were well, but not linearly correlated: VPAC(1) receptors activated G(alpha s) protein more efficiently but with the same pharmacological profile as the other G proteins. In contrast, there was a difference between cAMP increase in intact and broken cell membranes: EC(50) values were generally lower in intact cells whereas the efficacy was higher. There was, however, no correlation between the shift in the EC(50) value and the intrinsic activity. Of interest, the (4-28) fragment, a reported antagonist on cell membrane, was a full agonist in intact cells. We concluded that the active states of the VPAC(1) receptor resulting from the coupling to different effector are undistinguishable by the VIP analogs tested but that receptor properties are different when evaluated in intact cells or cell membranes.


Subject(s)
Receptors, Vasoactive Intestinal Peptide/agonists , Adenylyl Cyclases/metabolism , Animals , CHO Cells , Calcium/metabolism , Cell Membrane/metabolism , Cricetinae , Dose-Response Relationship, Drug , Receptors, Vasoactive Intestinal Polypeptide, Type I , Vasoactive Intestinal Peptide/pharmacology
9.
Mol Endocrinol ; 16(5): 1089-96, 2002 May.
Article in English | MEDLINE | ID: mdl-11981043

ABSTRACT

The stimulatory effect of VIP on intracellular calcium concentration ([Ca(2+)](i)) has been investigated in Chinese hamster ovary cells stably transfected with the reporter gene aequorin, and expressing human VPAC(1), VPAC(2), chimeric VPAC(1)/VPAC(2), or mutated receptors. The VIP-induced [Ca(2+)](i) increase was linearly correlated with receptor density and was higher in cells expressing VPAC(1) receptors than in cells expressing a similar VPAC(2) receptor density. The study was performed to establish the receptor sequence responsible for that difference. VPAC(1)/VPAC(2) chimeric receptors were first used for a broad positioning: those having the third intracellular loop (IC(3)) of the VPAC(1) or of the VPAC(2) receptor behaved, in that respect, phenotypically like VPAC(1) and VPAC(2) receptor, respectively. Replacement in the VPAC(2) receptor of the sequence 315-318 (VGGN) within the IC(3) by its VPAC(1) receptor counterpart 328-331 (IRKS) and the introduction of VGGN in state of IRKS in VPAC(1) was sufficient to mimic the VPAC(1) and VPAC(2) receptor characteristics, respectively. Thus, a small sequence in the IC(3) of the VPAC(1) receptor, probably through interaction with G(alphai) and G(alphaq) proteins, is responsible for the efficient agonist-stimulated [Ca(2+)](i) increase.


Subject(s)
Calcium/metabolism , Receptors, Vasoactive Intestinal Peptide/chemistry , Receptors, Vasoactive Intestinal Peptide/metabolism , Vasoactive Intestinal Peptide/metabolism , Vasoactive Intestinal Peptide/pharmacology , Adenylyl Cyclases/metabolism , Aequorin/genetics , Amino Acid Sequence , Animals , CHO Cells , Cricetinae , Cyclic AMP/metabolism , Dose-Response Relationship, Drug , Enzyme Activation , Gene Expression , Genes, Reporter , Humans , Molecular Sequence Data , Mutagenesis , Receptors, Vasoactive Intestinal Peptide/genetics , Receptors, Vasoactive Intestinal Polypeptide, Type I , Recombinant Fusion Proteins , Structure-Activity Relationship , Transfection
10.
Peptides ; 25(11): 1943-9, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15501526

ABSTRACT

C-terminally truncated human VPAC(1) receptors were constructed and stably transfected in Chinese hamster ovary (CHO) cells. Selected clones expressing comparable receptor densities were studied for ligand's binding properties, basal and stimulated adenylate cyclase activity. The wild-type (1-457) receptor served as reference. The binding properties of all the constructions were preserved. As judged by the intrinsic activity of the partial agonist Q(3)-VIP, the shortest receptors have a moderate impairment of the coupling efficacy to G(alpha s) protein. Cells expressing the VPAC(1) (1-436) and (1-441) truncated receptors had a two- to three-fold higher basal adenylate cyclase activity than those expressing the wild-type or the VPAC(1) (1-444), (1-433), (1-429), (1-421) and (1-398) receptor. The stimulatory effect of VIP and other agonist was preserved. This suggested that VPAC(1) (1-436) and (1-441) receptors had a constitutive activity. The selective VPAC(1) receptor antagonist Ac His(1) [D-Phe(2), K(15), R(16), L(27)] VIP (3-7)/GRF (8-27) reduced by 60% the basal activity with an EC(50) value of 3 nM comparable to its IC(50) value for binding. This agonist behaved thus like an inverse agonist on the constitutively active VPAC(1) receptors generated by C-terminal truncation and expressed in CHO cells.


Subject(s)
Receptors, Vasoactive Intestinal Peptide/chemistry , Vasoactive Intestinal Peptide/pharmacology , Adenylyl Cyclases/metabolism , Amino Acid Sequence , Animals , Binding, Competitive , CHO Cells , Clone Cells , Cricetinae , Cricetulus , Humans , Inhibitory Concentration 50 , Iodine Radioisotopes , Ligands , Molecular Sequence Data , Peptides, Cyclic/pharmacology , Protein Conformation , Receptors, Vasoactive Intestinal Peptide/metabolism , Receptors, Vasoactive Intestinal Polypeptide, Type I , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Transfection , Vasoactive Intestinal Peptide/chemical synthesis , Vasoactive Intestinal Peptide/chemistry , Vasoactive Intestinal Peptide/isolation & purification
11.
Peptides ; 25(2): 275-8, 2004 Feb.
Article in English | MEDLINE | ID: mdl-15063009

ABSTRACT

We synthesized a VIP analog that combines mutations that decrease the affinity for the VPAC1 receptor but maintain a high affinity for the VPAC2 receptor with an amino-terminal hexanoylation that increases the affinity for the VPAC2 receptor with a limited decrease in the affinity of the VPAC1 receptor. The resulting Hexanoyl[A19,K(27,28)]VIP had the expected properties of a high affinity for the VPAC2 receptor and a low affinity for the VPAC1 receptor and also a low affinity for the PAC1 and secretin receptors. With a 1000-fold preference for the VPAC2 receptor and a IC50 value of binding of 1 nM, this compound is the most potent and the most selective agonist presently described.


Subject(s)
Receptors, Gastrointestinal Hormone/agonists , Receptors, Vasoactive Intestinal Peptide/agonists , Vasoactive Intestinal Peptide/chemistry , Animals , Binding Sites , Humans , Protein Binding , Receptors, G-Protein-Coupled , Receptors, Vasoactive Intestinal Peptide, Type II , Receptors, Vasoactive Intestinal Polypeptide, Type I , Vasoactive Intestinal Peptide/analysis , Vasoactive Intestinal Peptide/chemical synthesis
12.
Eur J Pharmacol ; 478(1): 1-9, 2003 Sep 30.
Article in English | MEDLINE | ID: mdl-14555178

ABSTRACT

Levetiracetam (2S-(2-oxo-1-pyrrolidinyl)butanamide, KEPPRA, a novel antiepileptic drug, has been shown to bind to a specific binding site located in brain (levetiracetam binding site [Eur. J. Pharmacol. 286 (1995) 137]). However, [3H]levetiracetam displayed only micromolar affinity for these sites making it an unsuitable probe for further characterization. The present study describes the binding properties of an analogue of levetiracetam: [3H]ucb 30889, (2S)-2-[4-(3-azidophenyl)-2-oxopyrrolidin-1-yl]butanamide. [3H]ucb 30889 binds reversibly to specific binding sites in rat brain. Kinetics at 4 degrees C were biphasic with half-times of association and dissociation of, respectively, 3 and 4 min for the fast component and 47 and 61 min for the slow component. [3H]ucb 30889 saturation binding curves were compatible with the labelling of a homogenous population of binding sites having a B(max) of 4496+/-790 fmol/mg protein (mean+/-S.D., n=5) and a K(d) of 62+/-20 nM (mean+/-S.D., n=5), a 20-fold increase in affinity compared to [3H]levetiracetam. Competition binding curves with ligands known to interact with levetiracetam binding sites and tissue distribution restricted to the brain indicated that [3H]ucb 30889 and [3H]levetiracetam bind to the same site. Although levetiracetam binding sites and GABA(A) (gamma-aminobutyric acid) receptors share some ligands such as pentobarbital and pentylenetetrazol, experiments performed with [35S]TBPS (tert-butyl-bicyclo[2.2.2]phosporothionate), a probe for the GABA(A) Cl(-) channel do not support the hypothesis that levetiracetam binding sites are part of the GABA(A) receptor complex. Preliminary autoradiography studies in rat brain revealed that [3H]ucb 30889 labels specific sites in all brain regions and that this binding is concentration-dependently displaced by levetiracetam.


Subject(s)
Anticonvulsants/metabolism , Azides/metabolism , Brain/metabolism , Piracetam/metabolism , Pyrrolidines/metabolism , Animals , Anticonvulsants/chemistry , Azides/chemistry , Binding, Competitive/drug effects , Binding, Competitive/physiology , Brain/drug effects , Dose-Response Relationship, Drug , Levetiracetam , Male , Piracetam/analogs & derivatives , Piracetam/chemistry , Pyrrolidines/chemistry , Rats , Rats, Sprague-Dawley , Tissue Distribution/drug effects , Tissue Distribution/physiology , Tritium , gamma-Aminobutyric Acid/pharmacology
13.
Eur J Pharmacol ; 478(1): 11-9, 2003 Sep 30.
Article in English | MEDLINE | ID: mdl-14555179

ABSTRACT

Levetiracetam (2S-(2-oxo-1-pyrrolidinyl)butanamide, KEPPRA, a novel antiepileptic drug, has been shown to bind to a specific binding site located in the brain (Eur. J. Pharmacol. 286 (1995) 137). To identify the protein constituent of the levetiracetam binding site in situ, we synthesized the photoaffinity label [3H]ucb 30889 ((2S)-2-[4-(3-azidophenyl)-2-oxopyrrolidin-1-yl]butanamide), a levetiracetam analog with higher affinity for the levetiracetam binding site. This radioligand was used to map the levetiracetam binding site within the brain and to study its cellular and subcellular distribution. Autoradiography experiments using [3H]ucb 30889 in rat brain revealed a unique distribution profile that did not match that of classical receptors known to be involved in the generation of epileptic seizures. There was a high level of binding in the dentate gyrus, the superior colliculus, several thalamic nuclei, the molecular layer of the cerebellum and to a lesser extent in the cerebral cortex, the striatum and the hypothalamus. The levetiracetam binding site was restricted to neuronal cell types, undifferentiated PC12 cells and was highly enriched in synaptic vesicles. [3H]ucb 30889 was also used in photoaffinity labelling studies and shown to bind covalently to a membrane protein with a molecular weight of approximately 90 kDa.


Subject(s)
Brain/metabolism , Photoaffinity Labels/metabolism , Piracetam/metabolism , Animals , Binding Sites/physiology , Brain/cytology , COS Cells , Cell Line , Chlorocebus aethiops , Cricetinae , Dose-Response Relationship, Drug , Levetiracetam , Male , Mice , Neurons/metabolism , Piracetam/analogs & derivatives , Rats , Rats, Sprague-Dawley , Synaptic Vesicles/metabolism
14.
Peptides ; 32(8): 1593-9, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21704096

ABSTRACT

Glucagon is an important hormone for the prevention of hypoglycemia, and contributes to the hyperglycemia observed in diabetic patients, yet very little is known about its receptor structure and the receptor-glucagon interaction. In related receptors, the first extracellular loop, ECL1, is highly variable in length and sequence, suggesting that it might participate in ligand recognition. We applied a variant of the SCAM (Substituted Cysteine Accessibility Method) to the glucagon receptor ECL1 and sequentially mutated positions 197 to 223 to cysteine. Most of the mutations (15/27) affected the glucagon potency, due either to a modification of the glucagon binding site, or to the destabilization of the active receptor conformation. We reasoned that side chains accessible to glucagon must also be accessible to large, hydrophilic cysteine reagents. We therefore evaluated the accessibility of the introduced cysteines to maleimide-PEO(2)-biotin ((+)-biotinyl-3-maleimido-propionamidyl-3,6-dioxa-octanediamine), and tested the effect of pretreatment of intact cells with a large cationic cysteine reagent, MTSET ([2-(trimethylammonium)ethyl]methanethiosulfonate bromide), on glucagon potency. Our results suggest that the second and third transmembrane helices (TM2 and TM3) are extended to position 202 and from position 215, respectively, and separated by a short ß stretch (positions 203-209). Glucagon binding induced a conformational change close to TM2: L198C was accessible to the biotin reagent only in the presence of glucagon. Most other mutations affected the receptor activation rather than glucagon recognition, but S217 and D218 (at the top of TM3) were good candidates for glucagon recognition and V221 was very close to the binding site.


Subject(s)
Cysteine/genetics , Receptors, Glucagon/genetics , Amino Acid Sequence , Binding Sites , Cells, Cultured , Cysteine/metabolism , Glucagon/metabolism , Humans , Mesylates/pharmacology , Molecular Sequence Data , Mutagenesis, Site-Directed , Receptors, Glucagon/metabolism , Transfection
15.
J Biol Chem ; 280(30): 28034-43, 2005 Jul 29.
Article in English | MEDLINE | ID: mdl-15932876

ABSTRACT

When exposed to vasoactive intestinal peptide (VIP), the human wild type VPAC1 receptor expressed in Chinese hamster ovary (CHO) cells is rapidly phosphorylated, desensitized, and internalized in the endosomal compartment and is not re-expressed at the cell membrane within 2 h after agonist removal. The aims of the present work were first to correlate receptor phosphorylation level to internalization and recycling, measured by flow cytometry and in some cases by confocal microscopy using a monoclonal antibody that did not interfere with ligand binding, and second to identify the phosphorylated Ser/Thr residues. Combining receptor mutations and truncations allowed identification of Ser250 (in the second intracellular loop), Thr429, Ser435, Ser448 or Ser449, and Ser455 (all in the distal part of the C terminus) as candidates for VIP-stimulated phosphorylation. The effects of single mutations were not additive, suggesting alternative phosphorylation sites in mutated receptors. Replacement of all of the Ser/Thr residues in the carboxyl-terminal tail and truncation of the domain containing these residues completely inhibited VIP-stimulated phosphorylation and receptor internalization. There was, however, no direct correlation between receptor phosphorylation and internalization; in some truncated and mutated receptors, a 70% reduction in phosphorylation had little effect on internalization. In contrast to results obtained on the wild type and all of the mutated or truncated receptors that still underwent phosphorylation, internalization of the severely truncated receptor was reversed within 2 h of incubation in the absence of the agonist. Receptor recovery was blocked by monensin, an endosome inhibitor.


Subject(s)
Receptors, Vasoactive Intestinal Peptide/chemistry , Receptors, Vasoactive Intestinal Peptide/physiology , Adenylyl Cyclases/metabolism , Amino Acid Sequence , Animals , Antibodies, Monoclonal/chemistry , Blotting, Western , CHO Cells , Cell Line , Cell Membrane/metabolism , Cricetinae , Cyclic AMP/chemistry , Endosomes/metabolism , Flow Cytometry , Humans , Immunoprecipitation , Ligands , Microscopy, Confocal , Molecular Sequence Data , Monensin/chemistry , Mutation , Peptides/chemistry , Phosphorylation , Point Mutation , Protein Binding , Protein Structure, Tertiary , Protein Transport , Receptors, Vasoactive Intestinal Peptide/metabolism , Receptors, Vasoactive Intestinal Polypeptide, Type I , Serine/chemistry , Time Factors , Vasoactive Intestinal Peptide/chemistry
16.
Biochem J ; 370(Pt 3): 1003-9, 2003 Mar 15.
Article in English | MEDLINE | ID: mdl-12475394

ABSTRACT

We developed previously VPAC(1) [vasoactive intestinal peptide (VIP)/pituitary adenylate cyclase-activating peptide (PACAP) receptor]>VPAC(2) receptor selective ligands. Replacement of the VIP-Thr(11) by an Arg(11) in these ligands contributed to their selectivity: Arg(11)-VIP had a 200-fold lower affinity when compared with VIP at VPAC(2) receptors as opposed to 3- to 5-fold higher affinity at VPAC(1) receptors. Comparison of the binding and functional properties of related VIP analogues suggested that the VPAC(1) selectivity of Arg(11)-VIP was due to the loss of a hydrogen bond between the hydroxy group of Thr residue and the VPAC(2) receptor, steric hindrance between the Arg side chain and the VPAC(2) receptor and charge attraction by the VPAC(1) receptor. Comparison of the ability of VIP analogues to activate adenylate cyclase through chimaeric VPAC(1)/VPAC(2) and VPAC(2)/VPAC(1) receptors indicated that the first extracellular receptor loop carried most of the VPAC(2) receptors' ability to discriminate VIP from Arg(11)-VIP. Based on results obtained for a truncated VPAC(2) receptor and the closely related PACAP-preferring receptor (PAC(1)) and secretin receptors, we hypothesized that Thr(11) interacted with the VPAC(2) receptor Tyr(184) (similar to the VPAC(1) receptor Phe(200) residue). The Y184F (Tyr(184)-->Phe) VPAC(2) mutant lost the ability to discriminate VIP from Val(11)-VIP, and the F200Y VPAC(1) mutant acquired the ability to discriminate the natural peptide from Val(11)-VIP. These results support the hypothesis that the hydroxy group of the native VIP-Thr(11) side chain can indeed form a hydrogen bond with the Tyr side chain in the VPAC(2) receptor.


Subject(s)
Protein Conformation , Receptors, Vasoactive Intestinal Peptide/metabolism , Threonine/metabolism , Tyrosine/metabolism , Vasoactive Intestinal Peptide/chemistry , Vasoactive Intestinal Peptide/metabolism , Adenylyl Cyclases/metabolism , Cell Line , Cell Membrane/metabolism , Gastrointestinal Agents/chemistry , Gastrointestinal Agents/metabolism , Hydrogen Bonding , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Binding , Receptors, Vasoactive Intestinal Peptide/chemistry , Receptors, Vasoactive Intestinal Peptide/genetics , Receptors, Vasoactive Intestinal Peptide, Type II , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
17.
Biochem J ; 362(Pt 2): 389-94, 2002 Mar 01.
Article in English | MEDLINE | ID: mdl-11853547

ABSTRACT

Receptor recognition by the Asp(3) residues of vasoactive intestinal peptide and secretin requires the presence of a lysine residue close to the second transmembrane helix (TM2)/first extracellular loop junction and an ionic bond with an arginine residue in TM2. We tested whether the glucagon Gln(3) residue recognizes the equivalent positions in its receptor. Our data revealed that the binding and functional properties of the wild-type glucagon receptor and the K188R mutant were not significantly different, whereas all agonists had markedly lower potencies and affinities at the I195K mutated receptor. In contrast, glucagon was less potent and the Asp(3)-, Asn(3)- and Glu(3)-glucagon mutants were more potent and efficient at the double-mutated K188R/I195K receptor. Furthermore, these alterations were selective for position 3 of glucagon, as shown by the functional properties of the mutant Glu(9)- and Lys(15)-glucagon. Our results suggest that although the Gln(3) residue of glucagon did not interact with the equivalent binding pocket as the Asp(3) residue of vasoactive intestinal peptide or secretin, the Asp(3)-glucagon analogue was able to interact with position 188 of the K188R/I195K glucagon receptor. Nevertheless, the Gln(3) side chain of glucagon probably binds very close to this region in the wild-type receptor.


Subject(s)
Ligands , Receptors, Gastrointestinal Hormone/chemistry , Receptors, Glucagon/chemistry , Receptors, Pituitary Hormone/chemistry , Receptors, Vasoactive Intestinal Peptide/chemistry , Amino Acid Sequence , Animals , Binding Sites , CHO Cells , Cell Membrane/metabolism , Cricetinae , DNA Mutational Analysis , Kinetics , Molecular Sequence Data , Mutagenesis, Site-Directed , Rats , Receptors, G-Protein-Coupled , Receptors, Gastrointestinal Hormone/genetics , Receptors, Gastrointestinal Hormone/metabolism , Receptors, Glucagon/genetics , Receptors, Glucagon/metabolism , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide , Receptors, Pituitary Hormone/genetics , Receptors, Vasoactive Intestinal Peptide/genetics , Receptors, Vasoactive Intestinal Peptide/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL