Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Reproduction ; 162(6): 427-436, 2021 10 28.
Article in English | MEDLINE | ID: mdl-34605770

ABSTRACT

Polycystic ovarian syndrome (PCOS) is the main cause of infertility in women. It is frequently associated with reduced progesterone production by human luteinised granulosa cells (hlGCs). However, the molecular mechanisms involved in these steroidogenesis alterations in PCOS patients are unclear. In a dihydrotestosterone-induced PCOS mouse model, steroid production is maintained in the setting of chemokine-like receptor 1 (Cmklr1) knockout. Thus, chemerin and chemerin receptors in terms of expression and progesterone regulation could be different in control and PCOS hlGCs. We first confirmed that progesterone levels in both plasma (P < 0.0001) and follicular fluid (FF) (P < 0.0001) were significantly reduced in PCOS normal weight women compared to control women. These data were associated with a lower STAR mRNA expression in both in vivo (P < 0.0001) and in vitro (P < 0.0001) hlGCs from PCOS women. Secondly, chemerin FF levels (P < 0.0001) and RARRES2 (P < 0.05) and CMKLR1 (P < 0.0001) mRNA levels in GCs were higher in PCOS normal weight patients. Thirdly, treatment of hlGCs with a specific nanobody (the VHH CA4910) targeting the human receptor for CMKLR1 leading to its inactivation abolished chemerin-induced progesterone inhibition, suggesting the involvement of CMKLR1 in this process. Furthermore, the inhibition of progesterone secretion induced by chemerin was two-fold higher in PCOS hlGCs (P < 0.05). Moreover, the VHH CA4910 reinstated a normal progesterone secretion with lower concentrations in PCOS hlGCs, suggesting a different chemerin sensitivity between PCOS and control hlGCs. Thus, chemerin, through CMKLR1, could be involved in the steroidogenesis alterations in PCOS hlGCs.


Subject(s)
Chemokines/metabolism , Polycystic Ovary Syndrome , Animals , Chemokines/genetics , Female , Follicular Fluid/metabolism , Granulosa Cells/metabolism , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Polycystic Ovary Syndrome/metabolism , Progesterone/metabolism , Receptors, Chemokine/genetics , Receptors, Chemokine/metabolism
2.
Int J Mol Sci ; 20(18)2019 09 05.
Article in English | MEDLINE | ID: mdl-31491880

ABSTRACT

Vasoactive intestinal peptide receptor 1 (VPAC1) is a member of a secretin-like subfamily of G protein-coupled receptors. Its endogenous neuropeptide (VIP), secreted by neurons and immune cells, modulates various physiological functions such as exocrine and endocrine secretions, immune response, smooth muscles relaxation, vasodilation, and fetal development. As a drug target, VPAC1 has been selected for therapy of inflammatory diseases but drug discovery is still hampered by lack of its crystal structure. In this study we presented the homology model of this receptor constructed with the well-known web service GPCRM. The VPAC1 model is composed of extracellular and transmembrane domains that form a complex with an endogenous hormone VIP. Using the homology model of VPAC1 the mechanism of action of potential drug candidates for VPAC1 was described. Only two series of small-molecule antagonists of confirmed biological activity for VPAC1 have been described thus far. Molecular docking and a series of molecular dynamics simulations were performed to elucidate their binding to VPAC1 and resulting antagonist effect. The presented work provides the basis for the possible binding mode of VPAC1 antagonists and determinants of their molecular recognition in the context of other class B GPCRs. Until the crystal structure of VPAC1 will be released, the presented homology model of VPAC1 can serve as a scaffold for drug discovery studies and is available from the author upon request.


Subject(s)
Drug Design , Molecular Docking Simulation , Molecular Dynamics Simulation , Receptors, Vasoactive Intestinal Polypeptide, Type I/chemistry , Binding Sites , Humans , Ligands , Molecular Structure , Protein Binding , Protein Conformation , Quantitative Structure-Activity Relationship , Receptors, Vasoactive Intestinal Polypeptide, Type I/antagonists & inhibitors
3.
J Immunol ; 196(6): 2893-901, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26864035

ABSTRACT

The generation of Abs that recognize the native conformation of G protein-coupled receptors can be a challenging task because, like most multimembrane-spanning proteins, they are extremely difficult to purify as native protein. By combining genetic immunization, phage display, and biopanning, we identified two functional monovalent Abs (nanobodies) targeting ChemR23. The two nanobodies (CA4910 and CA5183) were highly specific for the human receptor and bind ChemR23 with moderate affinity. Binding studies also showed that they share a common binding site that overlaps with that of chemerin, the natural ligand of ChemR23. Consistent with these results, we found that the nanobodies were able to antagonize chemerin-induced intracellular calcium increase. The inhibition was partial when chemerin was used as agonist and complete when the chemerin(149-157) nonapeptide was used as agonist. Engineering of a bivalent CA4910 nanobody resulted in a relatively modest increase in affinity but a marked enhancement of efficacy as an antagonist of chemerin induced intracellular calcium mobilization and a much higher potency against the chemerin(149-157) nonapeptide-induced response. We also demonstrated that the fluorescently labeled nanobodies detect ChemR23 on the surface of human primary cell populations as efficiently as a reference mouse mAb and that the bivalent CA4910 nanobody behaves as an efficient antagonist of chemerin-induced chemotaxis of human primary cells. Thus, these nanobodies constitute new tools to study the role of the chemerin/ChemR23 system in physiological and pathological conditions.


Subject(s)
Dendritic Cells/metabolism , Macrophages/metabolism , Receptors, Chemokine/immunology , Single-Domain Antibodies/metabolism , Animals , Antibodies, Monoclonal/metabolism , Calcium Signaling , Camelids, New World , Cell Surface Display Techniques , Cells, Cultured , Chemokines/metabolism , DNA/administration & dosage , Genetic Engineering , Humans , Immunization , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Peptide Fragments/metabolism , Protein Binding , Single-Domain Antibodies/genetics , Single-Domain Antibodies/immunology
4.
Biomedicines ; 10(2)2022 Feb 09.
Article in English | MEDLINE | ID: mdl-35203615

ABSTRACT

Homeostasis of the human immune system is regulated by many cellular components, including two neuropeptides, VIP and PACAP, primary stimuli for three class B G protein-coupled receptors, VPAC1, VPAC2, and PAC1. Vasoactive intestinal peptide (VIP) and pituitary adenylate cyclase-activating polypeptide (PACAP) regulate intestinal motility and secretion and influence the functioning of the endocrine and immune systems. Inhibition of VIP and PACAP receptors is an emerging concept for new pharmacotherapies for chronic inflammation and cancer, while activation of their receptors provides neuroprotection. A small number of known active compounds for these receptors still impose limitations on their use in therapeutics. Recent cryo-EM structures of VPAC1 and PAC1 receptors in their agonist-bound active state have provided insights regarding their mechanism of activation. Here, we describe major molecular switches of VPAC1, VPAC2, and PAC1 that may act as triggers for receptor activation and compare them with similar non-covalent interactions changing upon activation that were observed for other GPCRs. Interhelical interactions in VIP and PACAP receptors that are important for agonist binding and/or activation provide a molecular basis for the design of novel selective drugs demonstrating anti-inflammatory, anti-cancer, and neuroprotective effects. The impact of genetic variants of VIP, PACAP, and their receptors on signalling mediated by endogenous agonists is also described. This sequence diversity resulting from gene splicing has a significant impact on agonist selectivity and potency as well as on the signalling properties of VIP and PACAP receptors.

5.
Front Endocrinol (Lausanne) ; 12: 711906, 2021.
Article in English | MEDLINE | ID: mdl-34867774

ABSTRACT

Vasoactive intestinal peptide (VIP) and pituitary adenylate cyclase-activating polypeptide (PACAP) are two neuropeptides that contribute to the regulation of intestinal motility and secretion, exocrine and endocrine secretions, and homeostasis of the immune system. Their biological effects are mediated by three receptors named VPAC1, VPAC2 and PAC1 that belong to class B GPCRs. VIP and PACAP receptors have been identified as potential therapeutic targets for the treatment of chronic inflammation, neurodegenerative diseases and cancer. However, pharmacological use of endogenous ligands for these receptors is limited by their lack of specificity (PACAP binds with high affinity to VPAC1, VPAC2 and PAC1 receptors while VIP recognizes both VPAC1 and VPAC2 receptors), their poor oral bioavailability (VIP and PACAP are 27- to 38-amino acid peptides) and their short half-life. Therefore, the development of non-peptidic small molecules or specific stabilized peptidic ligands is of high interest. Structural similarities between VIP and PACAP receptors are major causes of difficulties in the design of efficient and selective compounds that could be used as therapeutics. In this study we performed structure-based virtual screening against the subset of the ZINC15 drug library. This drug repositioning screen provided new applications for a known drug: ticagrelor, a P2Y12 purinergic receptor antagonist. Ticagrelor inhibits both VPAC1 and VPAC2 receptors which was confirmed in VIP-binding and calcium mobilization assays. A following analysis of detailed ticagrelor binding modes to all three VIP and PACAP receptors with molecular dynamics revealed its allosteric mechanism of action. Using a validated homology model of inactive VPAC1 and a recently released cryo-EM structure of active VPAC1 we described how ticagrelor could block conformational changes in the region of 'tyrosine toggle switch' required for the receptor activation. We also discuss possible modifications of ticagrelor comparing other P2Y12 antagonist - cangrelor, closely related to ticagrelor but not active for VPAC1/VPAC2. This comparison with inactive cangrelor could lead to further improvement of the ticagrelor activity and selectivity for VIP and PACAP receptor sub-types.


Subject(s)
Allosteric Regulation/drug effects , Drug Repositioning/methods , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/drug effects , Receptors, Vasoactive Intestinal Peptide, Type II/drug effects , Receptors, Vasoactive Intestinal Polypeptide, Type I/drug effects , Ticagrelor/pharmacology , Binding Sites , Computer Simulation , Drug Evaluation, Preclinical/methods , Molecular Structure , Protein Conformation/drug effects , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/chemistry , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Receptors, Vasoactive Intestinal Peptide, Type II/chemistry , Receptors, Vasoactive Intestinal Peptide, Type II/metabolism , Receptors, Vasoactive Intestinal Polypeptide, Type I/chemistry , Receptors, Vasoactive Intestinal Polypeptide, Type I/metabolism , Ticagrelor/chemistry
6.
Mol Pharmacol ; 78(3): 394-401, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20573782

ABSTRACT

The VPAC(1) receptor belongs to family B of G protein-coupled receptors (GPCR-B) and is activated upon binding of the vasoactive intestinal peptide (VIP). Despite the recent determination of the structure of the N terminus of several members of this receptor family, little is known about the structure of the transmembrane (TM) region and about the molecular mechanisms leading to activation. In the present study, we designed a new structural model of the TM domain and combined it with experimental mutagenesis experiments to investigate the interaction network that governs ligand binding and receptor activation. Our results suggest that this network involves the cluster of residues Arg(188) in TM2, Gln(380) in TM7, and Asn(229) in TM3. This cluster is expected to be altered upon VIP binding, because Arg(188) has been shown previously to interact with Asp(3) of VIP. Several point mutations at positions 188, 229, and 380 were experimentally characterized and were shown to severely affect VIP binding and/or VIP-mediated cAMP production. Double mutants built from reciprocal residue exchanges exhibit strong cooperative or anticooperative effects, thereby indicating the spatial proximity of residues Arg(188), Gln(380), and Asn(229). Because these residues are highly conserved in the GPCR-B family, they can moreover be expected to have a general role in mediating function.


Subject(s)
Receptors, Vasoactive Intestinal Polypeptide, Type I/metabolism , Animals , Asparagine/genetics , Asparagine/metabolism , Cellular Structures/metabolism , Cricetinae , Humans , Mutagenesis , Protein Structure, Secondary/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Vasoactive Intestinal Polypeptide, Type I/genetics , Vasoactive Intestinal Peptide/genetics , Vasoactive Intestinal Peptide/metabolism
7.
Mol Pharmacol ; 75(3): 502-13, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19064631

ABSTRACT

Signaling of G protein-coupled receptors (GPCRs) is regulated by different mechanisms. One of these involves regulators of G protein signaling (RGS), which are diverse and multifunctional proteins that bind to active Galpha subunits of G proteins and act as GTPase-activating proteins. Little is known about the molecular mechanisms that govern the selective use of RGS proteins in living cells. We first demonstrated that CCK2R-mediated inositol phosphate production, known to be G(q)-dependent, is more sensitive to RGS2 than to RGS4 and is insensitive to RGS8. Both basal and agonist-stimulated activities of the CCK2R are regulated by RGS2. By combining biochemical, functional, and in silico structural approaches, we demonstrate that a direct and functional interaction occurs between RGS2 and agonist-stimulated cholecystokinin receptor-2 (CCK2R) and identified the precise residues involved: phosphorylated Ser434 and Thr439 located in the C-terminal tail of CCK2R and Lys62, Lys63, and Gln67, located in the N-terminal domain of RGS2. These findings confirm previous reports that RGS proteins can interact with GPCRs to modulate their signaling and provide a molecular basis for RGS2 recognition by the CCK2R.


Subject(s)
Peptide Fragments/physiology , RGS Proteins/physiology , Receptor, Cholecystokinin B/physiology , Signal Transduction/physiology , Amino Acid Motifs/physiology , Amino Acid Sequence , Animals , COS Cells , Chlorocebus aethiops , Humans , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Phosphorylation/physiology , Protein Binding/physiology , RGS Proteins/chemistry , RGS Proteins/metabolism , Receptor, Cholecystokinin B/chemistry , Receptor, Cholecystokinin B/metabolism
8.
Biochim Biophys Acta ; 1783(9): 1663-72, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18435935

ABSTRACT

Like for most transmembrane proteins, translation of G protein-coupled receptors (GPCRs) mRNA takes place at the endoplasmic reticulum (ER) where they are synthesized, folded and assembled. The molecular mechanisms involved in the transport process of GPCRs from ER to the plasma membrane are poorly investigated. Here we studied the mechanisms involved in glycosylation-dependent cell surface expression and quality control of the receptor for Vasoactive Intestinal Polypeptide (VIP) VPAC1, a member of the B family of GPCRs. Using biochemical and pharmacological techniques and fluorescence microscopy, we have shown that only a fraction of newly synthesized VPAC1 attains properly conformation that allows their cell surface targeting. Misfolded or immature VPAC1 are taken in charge by co- and post-translational quality control that involves: 1) calnexin-dependent folding strictly through a glycan-dependent mechanism, 2) BiP-dependant folding, 3) translocation to the cytoplasm and proteasome-dependent degradation of improper proteins, and 4) post-ER quality control check points. Our data suggest that VPAC1 expression/trafficking pathways are under the control of complex and precise molecular mechanisms to ensure that only proper VPAC1 reaches the cell surface.


Subject(s)
Cell Membrane/metabolism , Proteasome Endopeptidase Complex/metabolism , Receptors, Vasoactive Intestinal Polypeptide, Type I/metabolism , Animals , CHO Cells , Cricetinae , Cricetulus , Molecular Chaperones/metabolism , Mutation , Protein Transport , Receptors, Vasoactive Intestinal Polypeptide, Type I/chemistry , Receptors, Vasoactive Intestinal Polypeptide, Type I/genetics , Temperature , Ubiquitin/metabolism
9.
Regul Pept ; 145(1-3): 17-23, 2008 Jan 10.
Article in English | MEDLINE | ID: mdl-17961734

ABSTRACT

CCK receptors represent potential targets in a number of diseases. Knowledge of CCK receptor binding sites is a prerequisite for the understanding of the molecular basis for their ligand recognition, partial agonism, ligand-induced trafficking of signalling. In the current paper, we report studies from our laboratory and others which have provided new data on the molecular basis of the pharmacology and functioning of CCK1 and CCK2 receptors. It has been shown that: 1) homologous regions of the two receptors are involved in the binding site of CCK, however, positioning of CCK slightly differs in agreement with distinct pharmacophores of CCK toward the two receptors and receptor sequence variations; 2) Binding sites of most of non-peptide agonists/ antagonist are buried in the pocket formed by transmembrane helices and overlap that of CCK; Aromatic amino acids within and near the binding site, especially in helix VI, are involved in receptor activation; 4) Like for other members of family A of G-protein coupled receptors, residues of the binding sites as well as of conserved motifs such as E/DRY, NPXXY are crucial for receptor activation.


Subject(s)
Cholecystokinin/metabolism , Gastrins/metabolism , Receptor, Cholecystokinin B/metabolism , Receptors, Cholecystokinin/metabolism , Animals , Binding Sites , Humans , Receptor, Cholecystokinin B/chemistry , Receptors, Cholecystokinin/chemistry
10.
Peptides ; 163: 170977, 2023 May.
Article in English | MEDLINE | ID: mdl-36804954
11.
Article in English | MEDLINE | ID: mdl-29674997

ABSTRACT

Multi-membrane spanning proteins, such as G protein-coupled receptors (GPCRs) and ion channels, are extremely difficult to purify as native proteins. Consequently, the generation of antibodies that recognize the native conformation can be challenging. By combining genetic immunization, phage display, and biopanning, we identified a panel of monovalent antibodies (nanobodies) targeting the vasoactive intestinal peptide receptor 1 (VPAC1) receptor. The nine unique nanobodies that were classified into four different families based on their CDR3 amino acid sequence and length, were highly specific for the human receptor and bind VPAC1 with moderate affinity. They all recognize a similar epitope localized in the extracellular N-terminal domain of the receptor and distinct from the orthosteric binding site. In agreement with binding studies, which showed that the nanobodies did not interfere with VIP binding, all nanobodies were devoid of any functional properties. However, we observed that the binding of two nanobodies was slightly increased in the presence of VPAC1 agonists [vasoactive intestinal polypeptide (VIP) and pituitary adenylate cyclase-activating polypeptide-27 (PACAP-27)], but decreased in the presence of VPAC1 antagonist. As no evidence of allosteric activity was seen in VIP binding studies nor in functional assays, it is, therefore, possible that the two nanobodies may behave as very weak allosteric modulators of VPAC1, detectable only in some sensitive settings, but not in others. We demonstrated that the fluorescently labeled nanobodies detect VPAC1 on the surface of human leukocytes as efficiently as a reference mouse monoclonal antibody. We also developed a protocol allowing efficient detection of VPAC1 by immunohistochemistry in paraffin-embedded human gastrointestinal tissue sections. Thus, these nanobodies constitute new original tools to further investigate the role of VPAC1 in physiological and pathological conditions.

12.
Cell Signal ; 18(12): 2121-30, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16650965

ABSTRACT

After stimulation with agonist, G protein coupled receptors (GPCR) undergo conformational changes that allow activation of G proteins to transduce the signal, followed by phosphorylation by kinases and arrestin binding to promote receptor internalization. Actual paradigm, based on a study of GPCR-A/rhodopsin family, suggests that a network of interactions between conserved residues located in transmembrane (TM) domains (mainly TM3, TM6 and TM7) is involved in the molecular switch leading to GPCR activation. We evaluated in CHO cells expressing the VPAC(1) receptor the role of the third transmembrane helix in agonist signalling by point mutation into Ala of the residues highly conserved in the secretin-family of receptors: Y(224), N(229), F(230), W(232), E(236), G(237), Y(239), L(240). N(229)A VPAC(1) mutant was characterized by a decrease in both potency and efficacy of VIP stimulated adenylate cyclase activity, by the absence of agonist stimulated [Ca(2+)](i) increase, by a preserved receptor recognition of agonists and antagonist and by a preserved sensitivity to GTP suggesting the importance of that residue for efficient G protein activation. N(229)D mutant was not expressed at the membrane, and the N(229)Q with a conserved mutation was less affected than the A mutant. Agonist stimulated phosphorylation and internalization of N(229)A and N(229)Q VPAC(1) were unaffected. However, the re-expression of internalized mutant receptors, but not that of the wild type receptor, was rapidly reversed after VIP washing. Receptor phosphorylation, internalization and re-expression may be thus dissociated from G protein activation and linked to another active conformation that may influence its trafficking. Mutation of that conserved amino acid in VPAC(2) could be investigated only by a conservative mutation (N(216)Q) and led to a receptor with a low VIP stimulation of adenylate cyclase, receptor phosphorylation and internalization. This indicated the importance of the conserved N residue in the TM3 of that family of receptors.


Subject(s)
Asparagine/metabolism , Endocytosis/physiology , Receptors, Vasoactive Intestinal Peptide, Type II/metabolism , Receptors, Vasoactive Intestinal Polypeptide, Type I/metabolism , Adenylyl Cyclases/metabolism , Amino Acid Sequence , Animals , Asparagine/genetics , Binding, Competitive , Biological Transport/physiology , CHO Cells , Calcium/metabolism , Cell Membrane/drug effects , Cell Membrane/metabolism , Cricetinae , Cricetulus , Enzyme Activation/drug effects , Kinetics , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation/genetics , Phosphorylation , Receptors, Vasoactive Intestinal Peptide, Type II/genetics , Receptors, Vasoactive Intestinal Polypeptide, Type I/genetics , Sequence Homology, Amino Acid , Transfection , Vasoactive Intestinal Peptide/analogs & derivatives , Vasoactive Intestinal Peptide/metabolism , Vasoactive Intestinal Peptide/pharmacology
13.
Molecules ; 12(7): 1290-306, 2007 Jul 05.
Article in English | MEDLINE | ID: mdl-17909485

ABSTRACT

Secondary plant compounds are important signals in several symbiotic and pathogenic plant-microbe interactions. The present review is limited to two groups of secondary plant compounds, flavonoids and strigolactones, which have been reported in root exudates. Data on flavonoids as signaling compounds are available from several symbiotic and pathogenic plant-microbe interactions, whereas only recently initial data on the role of strigolactones as plant signals in the arbuscular mycorrhizal symbiosis have been reported. Data from other plant-microbe interactions and strigolactones are not available yet. In the present article we are focusing on flavonoids in plant-fungal interactions such as the arbuscular mycorrhizal (AM) association and the signaling between different Fusarium species and plants. Moreover the role of strigolactones in the AM association is discussed and new data on the effect of strigolactones on fungi, apart from arbuscular mycorrhizal fungi (AMF), are provided.


Subject(s)
Flavonoids/metabolism , Fungi/physiology , Fungi/pathogenicity , Lactones/metabolism , Plant Exudates/chemistry , Plant Roots/metabolism , Plant Roots/microbiology , Symbiosis/physiology , Flavonoids/chemistry , Lactones/chemistry
14.
Br J Pharmacol ; 148(8): 1051-9, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16783404

ABSTRACT

1. In the light of recent findings that VPAC1 and VPAC2 receptors form homodimers and heterodimers, we have evaluated the function of these receptors coexpressed in the same cells, using whole-cell and membrane preparations. Cells expressing each receptor alone were used for comparison. 2. The study was performed on Chinese hamster ovary cells stably transfected with both human recombinant receptors and we compared receptor occupancy and adenylate cyclase activation by VIP, Ro 25-1553 - a VPAC2 selective agonist - and [K(15),R(16),L(27)]VIP(1-7)/GRF(8-27) - a VPAC1 selective agonist - on membranes prepared from each cell line and on a mixture of membranes from cells expressing each receptor individually. We also studied receptor internalization induced by the three agonists on intact cells expressing both receptors alone or together by fluorescence-activated cell sorting using monoclonal antibodies and demonstrated by using co-immunoprecipitation that the two receptors did interact.3. The results indicated that coexpression of the receptors did not modify the recognition of ligands, nor the capacity of the agonists to stimulate adenylate cyclase activity and, in intact cells, to induce internalization of the receptors.4. As a consequence, the properties of the selective ligands that were established on cell lines expressing a single population of VIP receptors were valid on cells expressing both receptors. Furthermore, the recently demonstrated VPAC1/VPAC2 receptor heterodimerization did not affect the function of either receptor.


Subject(s)
Receptors, Vasoactive Intestinal Peptide, Type II/metabolism , Receptors, Vasoactive Intestinal Polypeptide, Type I/metabolism , Adenylyl Cyclases/metabolism , Animals , Blotting, Western , CHO Cells , Cricetinae , Cricetulus , Endocytosis , Enzyme Activation , Humans , Immunoprecipitation , Radioligand Assay
15.
Peptides ; 27(7): 1865-70, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16554109

ABSTRACT

The hVPAC1 receptor is rapidly phosphorylated and internalized by agonists but not re-expressed at the membrane after washing. Mutation of Ser/Thr residues in the C-terminus reduced phosphorylation but not internalization that was abolished only when all the phosphorylatable residues were mutated. Substitution of Thr429 by Glu mimicking a phosphothreonin led to a mutant with unchanged binding properties, decreased coupling to adenylate cyclase consisting in a reduced VIP potency, increased basal and VIP stimulated phosphorylation, preserved internalization followed by a rapid receptor re-expression. These are the expected characteristics of a constitutively desensitized receptor, putting forward the role of Thr429 phosphorylation in that process.


Subject(s)
Glutamic Acid/chemistry , Mutation , Receptors, Vasoactive Intestinal Polypeptide, Type I/chemistry , Threonine/chemistry , Adenylyl Cyclases/metabolism , Animals , CHO Cells , Cricetinae , Humans , Peptides/chemistry , Phosphorylation , Protein Binding , Protein Structure, Tertiary
16.
Cell Signal ; 17(1): 17-24, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15451021

ABSTRACT

The vasoactive intestinal polypeptide (VIP) VPAC1 receptor is preferentially coupled to Galphas protein that stimulates adenylate cyclase activity and also to Galphaq and Galphai proteins that stimulate the inositol phosphate/calcium pathway. Previous studies indicated the importance of the third intracellular loop of the receptor for G protein coupling. By site-directed mutation of the human recombinant receptor expressed in Chinese hamster ovary cells, we identified two domains in this loop that contain clusters of basic residues conserved in most of the G-protein-coupled seven transmembrane domains receptors. We found that mutations in the proximal domain (K322) reduced the capability of VIP to increase adenylate cyclase activity without any change in the calcium response, whereas mutations in the distal part of the loop (R338, L339, R341) markedly reduced the calcium increase and Galphai coupling but only weakly the adenylate cyclase activity. Thus, the interaction of different G proteins with the VPAC1 receptor involves different receptor sub-domains.


Subject(s)
Adenylyl Cyclases/metabolism , Calcium/metabolism , Mutation , Receptors, Vasoactive Intestinal Peptide/chemistry , Receptors, Vasoactive Intestinal Peptide/genetics , Vasoactive Intestinal Peptide/pharmacology , Adenylyl Cyclases/drug effects , Amino Acid Sequence , Animals , Binding Sites , CHO Cells , Cricetinae , Humans , Molecular Sequence Data , Peptide Fragments , Protein Structure, Secondary , Receptors, Vasoactive Intestinal Polypeptide, Type I , Recombinant Proteins/chemistry
17.
Int J Pharm ; 501(1-2): 148-59, 2016 Mar 30.
Article in English | MEDLINE | ID: mdl-26850313

ABSTRACT

Despite the direct access to the lung offered by the inhalation route, drug penetration into lung tumors could remain an important issue. In this study, folate-polyethylene glycol-hydrophobically-modified dextran (F-PEG-HMD) micelles were developed as an effective pulmonary drug delivery system to reach and penetrate lung tumors and cancer cells. The F-PEG-HMD micelles were able to enter HeLa and M109-HiFR, two folate receptor-expressing cancer cell lines, in vitro, and in vivo after administration by inhalation to orthotopic M109-HiFR lung tumor grafted mice. Paclitaxel-loaded F-PEG-HMD micelles characterized in PBS by a Z-average diameter of ∼50 nm and a zeta potential of ∼-4 mV were prepared with an encapsulation efficiency of ∼100%. The loaded micelles reduced HeLa and M109-HiFR cell growth, with half maximal inhibitory concentrations of 37 and 150 nM, respectively. Dry powders embedding the paclitaxel-loaded F-PEG-HMD micelles were developed by spray-drying. In vitro, good deposition profiles were obtained, with a fine particle fraction of up to 50% and good ability to re-disperse the micelles in physiological buffer. A polymeric micelle-based dry powder without paclitaxel was well-tolerated in vivo, as assessed in healthy mice by determination of total protein content, cell count, and cytokine IL-1ß, IL-6, and TNF-α concentrations in bronchoalveolar lavage fluids.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Lung Neoplasms/drug therapy , Paclitaxel/administration & dosage , Administration, Inhalation , Animals , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/therapeutic use , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Cell Count , Cell Line, Tumor , Cell Survival/drug effects , Cytokines/metabolism , Dextrans/chemistry , Drug Liberation , Female , Folic Acid/analogs & derivatives , Folic Acid/chemistry , Humans , Leucine/chemistry , Lung Neoplasms/metabolism , Mannitol/chemistry , Mice , Mice, Inbred BALB C , Micelles , Paclitaxel/chemistry , Paclitaxel/therapeutic use , Polyethylene Glycols/chemistry , Powders
18.
ACS Synth Biol ; 5(10): 1070-1075, 2016 10 21.
Article in English | MEDLINE | ID: mdl-27176489

ABSTRACT

Membrane protein research is still hampered by the generally very low levels at which these proteins are naturally expressed, necessitating heterologous expression. Protein degradation, folding problems, and undesired post-translational modifications often occur, together resulting in low expression levels of heterogeneous protein products that are unsuitable for structural studies. We here demonstrate how the integration of multiple engineering modules in Pichia pastoris can be used to increase both the quality and the quantity of overexpressed integral membrane proteins, with the human CXCR4 G-protein coupled receptor as an example. The combination of reduced proteolysis, enhanced ER folding capacity, GlycoDelete-based N-Glycan trimming, and nanobody-based fold stabilization improved the expression of this GPCR in P. pastoris from a low expression level of a heterogeneously glycosylated, proteolyzed product to substantial quantities (2-3 mg/L shake flask culture) of a nonproteolyzed, homogeneously glycosylated proteoform. We expect that this set of tools will contribute to successful expression of more membrane proteins in a quantity and quality suitable for functional and structural studies.


Subject(s)
Genetic Engineering/methods , Pichia/genetics , Receptors, G-Protein-Coupled/genetics , Recombinant Proteins/genetics , Animals , CHO Cells , Camelids, New World , Cricetulus , Gene Library , Glycosylation , Metabolic Engineering/methods , Pichia/metabolism , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Receptors, G-Protein-Coupled/metabolism , Recombinant Proteins/metabolism , Single-Domain Antibodies/metabolism , Unfolded Protein Response/genetics
19.
J Mol Endocrinol ; 34(2): 405-14, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15821106

ABSTRACT

The VPAC(2) receptor, as all members of the G-protein-coupled receptor (GPCR)-B family, has two highly conserved motifs in the third intracellular (IC(3)) loop: a lysine and a leucine located at the amino-terminus and two basic residues separated by a leucine and an alanine at the carboxyl-terminus. This study evaluates the involvement of those conserved amino acid sequences in VPAC(2) signal transduction and regulation. The residues were mutated into alanine and mutants were expressed in Chinese hamster ovary (CHO) cells stably transfected with Galpha16 and aequorin. Mutation of L310 reduced efficacy of vasoactive intestinal polypeptide (VIP) to stimulate adenylate cyclase activity through Galphas coupling by 75%, without affecting VIP capability to stimulate an increase in [Ca(2+)](i) through Galpha16 coupling. Mutation of R325 and, to a lesser extend, K328 reduced VIP efficacy to stimulate [Ca(2+)](i) increase and VIP potency to stimulate adenylate cyclase. The combination of mutations of both amino- and carboxyl-terminus located conserved motifs of the IC(3) loop generates an inactive receptor with respect to [Ca(2+)](i) increase and adenylate cyclase activation, but also with respect to receptor phosphorylation and internalization that were indeed directly correlated with the potency of inactivation of the receptors. The amino-terminus of the VPAC(2) receptor IC(3) loop is thus involved in adenylate cyclase activation and the carboxyl-terminus of the IC(3) loop participates in both Galphas and Galpha16 coupling. The mutations studied also reduced both receptor phosphorylation and internalization in a manner that appeared directly linked to the alteration of Galphas and Galpha16 coupling.


Subject(s)
Endocytosis/physiology , Mutation , Receptors, Vasoactive Intestinal Peptide/genetics , Receptors, Vasoactive Intestinal Peptide/metabolism , Adenylyl Cyclases/metabolism , Amino Acid Sequence , Animals , CHO Cells , Calcium/metabolism , Cricetinae , Cyclic AMP/analogs & derivatives , Cyclic AMP/metabolism , Enzyme Activation , Enzyme Inhibitors/metabolism , Humans , Molecular Sequence Data , Phosphorylation , Protein Structure, Tertiary , Receptors, Vasoactive Intestinal Peptide/agonists , Receptors, Vasoactive Intestinal Peptide/chemistry , Receptors, Vasoactive Intestinal Peptide, Type II , Vasoactive Intestinal Peptide/metabolism
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL