Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters











Publication year range
1.
Mol Cell Endocrinol ; 589: 112235, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38621656

ABSTRACT

Luteinizing hormone (LH) is essential for reproduction, controlling ovulation and steroidogenesis. Its receptor (LHR) recruits various transducers leading to the activation of a complex signaling network. We recently identified iPRC1, the first variable fragment from heavy-chain-only antibody (VHH) interacting with intracellular loop 3 (ICL3) of the follicle-stimulating hormone receptor (FSHR). Because of the high sequence similarity of the human FSHR and LHR (LHCGR), here we examined the ability of the iPRC1 intra-VHH to modulate LHCGR activity. In this study, we demonstrated that iPRC1 binds LHCGR, to a greater extent when the receptor was stimulated by the hormone. In addition, it decreased LH-induced cAMP production, cAMP-responsive element-dependent transcription, progesterone and testosterone production. These impairments are not due to Gs nor ß-arrestin recruitment to the LHCGR. Consequently, iPRC1 is the first intra-VHH to bind and modulate LHCGR biological activity, including steroidogenesis. It should help further understand signaling mechanisms elicited at this receptor and their outcomes on reproduction.


Subject(s)
Luteinizing Hormone , Receptors, LH , Signal Transduction , Receptors, LH/metabolism , Receptors, LH/genetics , Humans , Signal Transduction/drug effects , Luteinizing Hormone/metabolism , Animals , Cyclic AMP/metabolism , Protein Binding , Progesterone/metabolism , Receptors, FSH/metabolism , Receptors, FSH/genetics , Testosterone/metabolism , Testosterone/biosynthesis , HEK293 Cells , GTP-Binding Proteins/metabolism , Steroids/biosynthesis , Steroids/metabolism
2.
FEBS Lett ; 598(2): 220-232, 2024 01.
Article in English | MEDLINE | ID: mdl-37923554

ABSTRACT

Intracellular variable fragments of heavy-chain antibody from camelids (intra-VHH) have been successfully used as chaperones to solve the 3D structure of active G protein-coupled receptors bound to their transducers. However, their effect on signalling has been poorly explored, although they may provide a better understanding of the relationships between receptor conformation and activity. Here, we isolated and characterized iPRC1, the first intra-VHH recognizing a member of the large glycoprotein hormone receptor family, the follicle-stimulating hormone receptor (FSHR). This intra-VHH recognizes the FSHR third intracellular loop and decreases cAMP production in response to FSH, without altering Gαs recruitment. Hence, iPRC1 behaves as an allosteric modulator and provides a new tool to complete structure/activity studies performed thus far on this receptor.


Subject(s)
Follicle Stimulating Hormone , Receptors, FSH , Receptors, FSH/genetics , Receptors, FSH/chemistry , Receptors, FSH/metabolism , Follicle Stimulating Hormone/chemistry , Follicle Stimulating Hormone/metabolism , Follicle Stimulating Hormone/pharmacology , GTP-Binding Proteins/metabolism , Signal Transduction
3.
Int J Mol Sci ; 24(21)2023 Nov 04.
Article in English | MEDLINE | ID: mdl-37958944

ABSTRACT

Developing modulatory antibodies against G protein-coupled receptors is challenging. In this study, we targeted the follicle-stimulating hormone receptor (FSHR), a significant regulator of reproduction, with variable domains of heavy chain-only antibodies (VHHs). We built two immune VHH libraries and submitted them to multiplexed phage display approaches. We used next-generation sequencing to identify 34 clusters of specifically enriched sequences that were functionally assessed in a primary screen based on a cAMP response element (CRE)-dependent reporter gene assay. In this assay, 23 VHHs displayed negative or positive modulation of FSH-induced responses, suggesting a high success rate of the multiplexed strategy. We then focused on the largest cluster identified (i.e., PRC1) that displayed positive modulation of FSH action. We demonstrated that PRC1 specifically binds to the human FSHR and human FSHR/FSH complex while potentiating FSH-induced cAMP production and Gs recruitment. We conclude that the improved selection strategy reported here is effective for rapidly identifying functionally active VHHs and could be adapted to target other challenging membrane receptors. This study also led to the identification of PRC1, the first potential positive modulator VHH reported for the human FSHR.


Subject(s)
Bacteriophages , Receptors, FSH , Humans , Receptors, FSH/genetics , Receptors, FSH/metabolism , Follicle Stimulating Hormone/metabolism , Signal Transduction , High-Throughput Nucleotide Sequencing , Bacteriophages/genetics
4.
Int J Mol Sci ; 23(17)2022 Aug 28.
Article in English | MEDLINE | ID: mdl-36077163

ABSTRACT

Developing a therapeutic antibody is a long, tedious, and expensive process. Many obstacles need to be overcome, such as biophysical properties (issues of solubility, stability, weak production yields, etc.), as well as cross-reactivity and subsequent toxicity, which are major issues. No in silico method exists today to solve such issues. We hypothesized that if we were able to properly measure the similarity between the CDRs of antibodies (Ab) by considering not only their evolutionary proximity (sequence identity) but also their structural features, we would be able to identify families of Ab recognizing similar epitopes. As a consequence, Ab within the family would share the property to recognize their targets, which would allow (i) to identify off-targets and forecast the cross-reactions, and (ii) to identify new Ab specific for a given target. Testing our method on 238D2, an antagonistic anti-CXCR4 nanobody, we were able to find new nanobodies against CXCR4 and to identify influenza hemagglutinin as an off-target of 238D2.


Subject(s)
Influenza, Human , Single-Domain Antibodies , Antibodies , Epitopes , Hemagglutinins , Humans
5.
Methods Mol Biol ; 1957: 177-194, 2019.
Article in English | MEDLINE | ID: mdl-30919355

ABSTRACT

ß-arrestins are so-called hub proteins: they make complexes with many different partners, assembling functional complexes, and thereby fulfilling their biological function. The importance of this process in G protein-coupled receptor (GPCR) signalling has been fully demonstrated for many different receptors. For direct interactions, determining the interface regions, on ß-arrestins and on the partners, is crucial for understanding the function of the complex. Indeed, this brings information on which proteins can interact simultaneously with ß-arrestins, or, on the contrary, which partners are exclusive. We present here a method in two steps: protein-protein docking allows finding a limited number of peptides predicted to be involved in the interaction, and then experimental approaches that might be used for validating the prediction.


Subject(s)
Molecular Biology/methods , beta-Arrestins/metabolism , Amino Acid Sequence , Fluorescence , Humans , Interferometry , Peptides/chemistry , Peptides/metabolism , Protein Binding , Reproducibility of Results , beta-Arrestins/chemistry
6.
J Immunol ; 201(10): 3096-3105, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30322966

ABSTRACT

Abs are very efficient drugs, ∼70 of them are already approved for medical use, over 500 are in clinical development, and many more are in preclinical development. One important step in the characterization and protection of a therapeutic Ab is the determination of its cognate epitope. The gold standard is the three-dimensional structure of the Ab/Ag complex by crystallography or nuclear magnetic resonance spectroscopy. However, it remains a tedious task, and its outcome is uncertain. We have developed MAbTope, a docking-based prediction method of the epitope associated with straightforward experimental validation procedures. We show that MAbTope predicts the correct epitope for each of 129 tested examples of Ab/Ag complexes of known structure. We further validated this method through the successful determination, and experimental validation (using human embryonic kidney cells 293), of the epitopes recognized by two therapeutic Abs targeting TNF-α: certolizumab and golimumab.


Subject(s)
Antibodies, Monoclonal/chemistry , Epitope Mapping/methods , Molecular Docking Simulation/methods , HEK293 Cells , Humans
7.
FASEB J ; 32(3): 1154-1169, 2018 03.
Article in English | MEDLINE | ID: mdl-29084767

ABSTRACT

Many interaction partners of ß-arrestins intervene in the control of mRNA translation. However, how ß-arrestins regulate this cellular process has been poorly explored. In this study, we show that ß-arrestins constitutively assemble a p70S6K/ribosomal protein S6 (rpS6) complex in HEK293 cells and in primary Sertoli cells of the testis. We demonstrate that this interaction is direct, and experimentally validate the interaction interface between ß-arrestin 1 and p70S6K predicted by our docking algorithm. Like most GPCRs, the biological function of follicle-stimulating hormone receptor (FSHR) is transduced by G proteins and ß-arrestins. Upon follicle-stimulating hormone (FSH) stimulation, activation of G protein-dependent signaling enhances p70S6K activity within the ß-arrestin/p70S6K/rpS6 preassembled complex, which is not recruited to the FSHR. In agreement, FSH-induced rpS6 phosphorylation within the ß-arrestin scaffold was decreased in cells depleted of Gαs. Integration of the cooperative action of ß-arrestin and G proteins led to the translation of 5' oligopyrimidine track mRNA with high efficacy within minutes of FSH input. Hence, this work highlights new relationships between G proteins and ß-arrestins when acting cooperatively on a common signaling pathway, contrasting with their previously shown parallel action on the ERK MAP kinase pathway. In addition, this study provides insights into how GPCR can exert trophic effects in the cell.-Tréfier, A., Musnier, A., Landomiel, F., Bourquard, T., Boulo, T., Ayoub, M. A., León, K., Bruneau, G., Chevalier, M., Durand, G., Blache, M.-C., Inoue, A., Fontaine, J., Gauthier, C., Tesseraud, S., Reiter, E., Poupon, A., Crépieux, P. G protein-dependent signaling triggers a ß-arrestin-scaffolded p70S6K/ rpS6 module that controls 5'TOP mRNA translation.


Subject(s)
5' Untranslated Regions/genetics , GTP-Binding Proteins/metabolism , Protein Biosynthesis , RNA, Messenger/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Ribosomal Protein S6/metabolism , beta-Arrestins/metabolism , Animals , Male , Protein Interaction Maps , RNA Processing, Post-Transcriptional , RNA, Messenger/genetics , Rats , Rats, Wistar , Receptors, FSH/metabolism , Sertoli Cells/metabolism , Signal Transduction
8.
Mol Hum Reprod ; 23(10): 685-697, 2017 10 01.
Article in English | MEDLINE | ID: mdl-29044421

ABSTRACT

STUDY QUESTION: Are four urinary hCG/menotropin (hMG) and one recombinant preparation characterized by different molecular features and do they mediate specific intracellular signaling and steroidogenesis? SUMMARY ANSWER: hCG and hMG preparations have heterogeneous compositions and mediate preparation-specific cell signaling and early steroidogenesis, although similar progesterone plateau levels are achieved in 24 h-treated human primary granulosa cells in vitro. WHAT IS KNOWN ALREADY: hCG is the pregnancy hormone marketed as a drug for ARTs to induce final oocyte maturation and ovulation, and to support FSH action. Several hCG formulations are commercially available, differing in source, purification methods and biochemical composition. STUDY DESIGN, SIZE, DURATION: Commercial hCG preparations for ART or research purposes were compared in vitro. PARTICIPANTS/MATERIALS, SETTING, METHODS: The different preparations were quantified by immunoassay with calibration against the hCG standard (Fifth IS; NIBSC 07/364). Immunoreactivity patterns, isoelectric points and oligosaccharide contents of hCGs were evaluated using reducing and non-reducing Western blotting, capillary isoelectric-focusing immunoassay and lectin-ELISA, respectively. Functional studies were performed in order to evaluate intracellular and total cAMP, progesterone production and ß-arrestin 2 recruitment by ELISA and BRET, in both human primary granulosa lutein cells (hGLC) and luteinizing hormone (LH)/hCG receptor (LHCGR)-transfected HEK293 cells, stimulated by increasing hormone concentrations. Statistical analysis was performed using two-way ANOVA and Bonferroni post-test or Mann-Whitney's U-test as appropriate. MAIN RESULTS AND THE ROLE OF CHANCE: Heterogeneous profiles were found among preparations, revealing specific molecular weight patterns (20-75 KDa range), isoelectric points (4.0-9.0 pI range) and lectin binding (P < 0.05; n = 7-10). These drug-specific compositions were linked to different potencies on cAMP production (EC50 1.0-400.0 ng/ml range) and ß-arrestin 2 recruitment (EC50 0.03-2.0 µg/ml) in hGLC and transfected HEK293 cells (P < 0.05; n = 3-5). In hGLC, these differences were reflected by preparation-specific 8-h progesterone production although similar plateau levels of progesterone were acheived by 24-h treatment (P ≥ 0.05; n = 3). LARGE SCALE DATA: N/A. LIMITATIONS, REASONS FOR CAUTION: The biological activity of commercial hCG/hMG preparations is provided in International Units (IU) by in-vivo bioassay and calibration against an International Standard, although it is an unsuitable unit of measure for in-vitro studies. The re-calibration against recombinant hCG,quantified in grams, is based on the assumption that all of the isoforms and glycosylation variants have similar immunoreactivity. WIDER IMPLICATIONS OF THE FINDINGS: hCG/hMG preparation-specific cell responses in vitro may be proposed to ART patients affected by peculiar ovarian response, such as that caused by polycystic ovary syndrome. Otherwise, all the preparations available for ART may provide a similar clinical outcome in healthy women. STUDY FUNDING AND COMPETING INTEREST(S): This study was supported by a grant of the Italian Ministry of Education, University and Research (PRIN 2015XCR88M). The authors have no conflict of interest.


Subject(s)
Chorionic Gonadotropin/chemistry , Fertility Agents, Female/chemistry , Granulosa Cells/drug effects , Menotropins/chemistry , Progesterone/biosynthesis , Signal Transduction/drug effects , Adult , Chorionic Gonadotropin/pharmacology , Cyclic AMP/biosynthesis , Female , Fertility Agents, Female/pharmacology , Follicle Stimulating Hormone/genetics , Follicle Stimulating Hormone/metabolism , Gene Expression Regulation , Granulosa Cells/cytology , Granulosa Cells/metabolism , HEK293 Cells , Humans , Isoelectric Point , Luteal Phase/physiology , Menotropins/pharmacology , Molecular Weight , Ovulation Induction/methods , Pregnancy , Primary Cell Culture , Receptors, LH/genetics , Receptors, LH/metabolism , Transfection , beta-Arrestin 2/genetics , beta-Arrestin 2/metabolism
9.
J Mol Endocrinol ; 52(3): 373-82, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24711644

ABSTRACT

The control of mRNA translation has been mainly explored in response to activated tyrosine kinase receptors. In contrast, mechanistic details on the translational machinery are far less available in the case of ligand-bound G protein-coupled receptors (GPCRs). In this study, using the FSH receptor (FSH-R) as a model receptor, we demonstrate that part of the translational regulations occurs by phosphorylation of the translation pre-initiation complex scaffold protein, eukaryotic initiation factor 4G (eIF4G), in HEK293 cells stably expressing the FSH-R. This phosphorylation event occurred when eIF4G was bound to the mRNA 5' cap, and probably involves mammalian target of rapamycin. This regulation might contribute to cap-dependent translation in response to FSH. The cap-binding protein eIF4E also had its phosphorylation level enhanced upon FSH stimulation. We also show that FSH-induced signaling not only led to cap-dependent translation but also to internal ribosome entry site (IRES)-dependent translation of some mRNA. These data add detailed information on the molecular bases underlying the regulation of selective mRNA translation by a GPCR, and a topological model recapitulating these mechanisms is proposed.


Subject(s)
Eukaryotic Initiation Factor-4G/metabolism , Follicle Stimulating Hormone/metabolism , Protein Biosynthesis/genetics , Receptors, FSH/genetics , Adaptor Proteins, Signal Transducing/metabolism , Cell Cycle Proteins , Cell Line, Tumor , Enzyme Activation , Gene Expression Regulation/genetics , HEK293 Cells , Humans , Peptide Chain Initiation, Translational/genetics , Peptide Initiation Factors , Phosphoproteins/metabolism , Phosphorylation , RNA, Messenger/genetics , Receptors, FSH/biosynthesis , Receptors, FSH/metabolism , Ribosomes/genetics , TOR Serine-Threonine Kinases
10.
Mol Endocrinol ; 26(4): 669-80, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22383463

ABSTRACT

FSH is a key hormonal regulator of Sertoli cell secretory activity, required to optimize sperm production. To fulfil its biological function, FSH binds a G protein-coupled receptor, the FSH-R. The FSH-R-transduced signaling network ultimately leads to the transcription or down-regulation of numerous genes. In addition, recent evidence has suggested that FSH might also regulate protein translation. However, this point has never been demonstrated conclusively yet. Here we have addressed this issue in primary rat Sertoli cells endogenously expressing physiological levels of FSH-R. We observed that, within 90 min of stimulation, FSH not only enhanced overall protein synthesis in a mammalian target of rapamycin-dependent manner but also increased the recruitment of mRNA to polysomes. m(7)GTP pull-down experiments revealed the functional recruitment of mammalian target of rapamycin and p70 S6 kinase to the 5'cap, further supported by the enhanced phosphorylation of one of p70 S6 kinase targets, the eukaryotic initiation factor 4B. Importantly, the scaffolding eukaryotic initiation factor 4G was also recruited, whereas eukaryotic initiation factor 4E-binding protein, the eukaryotic initiation factor 4E generic inhibitor, appeared to play a minor role in translational regulations induced by FSH, in contrast to what is generally observed in response to anabolic factors. This particular regulation of the translational machinery by FSH stimulation might support mRNA-selective translation, as shown here by quantitative RT-PCR amplification of the c-fos and vascular endothelial growth factor mRNA but not of all FSH target mRNA, in polysomal fractions. These findings add a new level of complexity to FSH biological roles in its natural target cells, which has been underappreciated so far.


Subject(s)
Follicle Stimulating Hormone/physiology , Protein Biosynthesis , RNA, Messenger/metabolism , Sertoli Cells/metabolism , Animals , Carrier Proteins/metabolism , Cells, Cultured , Eukaryotic Initiation Factor-4G/metabolism , Intracellular Signaling Peptides and Proteins , Male , Phosphoproteins/metabolism , Phosphorylation , Polyribosomes/metabolism , Primary Cell Culture , Proto-Oncogene Proteins c-fos/metabolism , RNA Caps/genetics , RNA Caps/metabolism , RNA, Messenger/genetics , Rats , Rats, Wistar , Receptors, FSH/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , TOR Serine-Threonine Kinases/metabolism
11.
Mol Cell Endocrinol ; 315(1-2): 271-6, 2010 Feb 05.
Article in English | MEDLINE | ID: mdl-19778579

ABSTRACT

Follicle-stimulating hormone (FSH) controls the proliferation and differentiation of Sertoli cells of the testis. FSH binds a G protein-coupled receptor (GPCR) to stimulate downstream effectors of the phosphoinositide-3 kinase (PI3K)-dependent pathway, without enhancing PI3K activity. To clarify this paradox, we explored the activity of phosphatase and tensin homolog deleted in chromosome 10 (PTEN), the PI3K major regulator, in primary cultures of rat Sertoli cells. We show that, within minutes, FSH increases PTEN neo-synthesis, requiring the proteasomal degradation of an unidentified intermediate, as well as PTEN enzymatic activity. Importantly, introducing an antisense cDNA of PTEN into differentiating Sertoli cells restores FSH-dependent cell proliferation. In conclusion, these results provide a new mechanism of PTEN regulation, which could serve to block entry into S phase of Sertoli cells, while they are proceeding through differentiation in prepubertal animals.


Subject(s)
Follicle Stimulating Hormone/pharmacology , Mitosis/drug effects , PTEN Phosphohydrolase/metabolism , Puberty/physiology , Sertoli Cells/drug effects , Sertoli Cells/physiology , Animals , Cells, Cultured , Follicle Stimulating Hormone/metabolism , Humans , Male , Mitosis/physiology , PTEN Phosphohydrolase/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositols/metabolism , Rats , Rats, Wistar , Sertoli Cells/cytology , Swine , Transferrin/metabolism
12.
Cell Mol Life Sci ; 66(21): 3487-503, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19730801

ABSTRACT

The mechanisms whereby G protein-coupled receptors (GPCR) activate signalling pathways involved in mRNA translation are ill-defined, in contrast to tyrosine kinase receptors (TKR). We compared a GPCR and a TKR, both endogenously expressed, for their ability to mediate phosphorylation of 70-kDa ribosomal S6 kinase p70S6K in primary rat Sertoli cells at two developmental stages. In proliferating cells stimulated with follicle-stimulating hormone (FSH), active p70S6K was phosphorylated on T389 and T421/S424, through cAMP-dependent kinase (PKA) and phosphatidyl-inositide-3 kinase (PI3K) antagonizing actions. In FSH-stimulated differentiating cells, active p70S6K was phosphorylated solely on T389, PKA and PI3K independently enhancing its activity. At both developmental stages, insulin-induced p70S6K regulation was consistent with reported data. Therefore, TKR and GPCR trigger distinct p70S6K active conformations. p70S6K developmental regulation was formalized in a dynamic mathematical model fitting the data, which led to experimentally inaccessible predictions on p70S6K phosphorylation rate.


Subject(s)
Cell Differentiation/physiology , Models, Biological , Receptors, G-Protein-Coupled/physiology , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Sertoli Cells/physiology , Animals , Cell Culture Techniques , Cell Differentiation/drug effects , Cells, Cultured , Chromones/pharmacology , Computer Simulation , Cyclic AMP-Dependent Protein Kinases/metabolism , Follicle Stimulating Hormone/pharmacology , Male , Morpholines/pharmacology , Phosphorylation/drug effects , Protein Conformation , Protein Kinase Inhibitors/pharmacology , Rats , Rats, Wistar , Receptors, G-Protein-Coupled/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/chemistry , Sertoli Cells/drug effects , Sertoli Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL