Your browser does not support JavaScript!

Home    Διαλεύκανση της δομής της τρίτης διαμεμβρανικής περιοχής του τύπου 1 υποδοχέα του CRF, καθώς και των αλληλεπιδράσεων του με τον CRF και των αναλόγων του.  

Results - Details

Add to Basket
[Add to Basket]
Identifier 000404134
Title Διαλεύκανση της δομής της τρίτης διαμεμβρανικής περιοχής του τύπου 1 υποδοχέα του CRF, καθώς και των αλληλεπιδράσεων του με τον CRF και των αναλόγων του.
Author Σπυριδάκη Κατερίνα
Thesis advisor Λιαπάκης, Γεώργιος
Reviewer Γραβάνης, Α.
Μαργιωρής, Α.
Θερμού, Κ
Μαυρομούστακος, Θ.
Βενυχάκη, Μ.
Χαραλαμπόπουλος, Ι.
Abstract The type 1 receptor (CRF1R) for the corticotropin-releasing factor (CRF) is a family B G protein coupled receptor (GPCR) (1), which plays a key role in the maintenance of homeostasis by regulating neural and endocrine functions (2,3). Malfunction of CRF/CRF1R systems is related with several diseases such as depression and anxiety. The clinical importance of CRF1R is further supported by that CRF1R-selective non-peptide small-molecule antagonists have been shown to display anxiolytic and antidepressant properties in animal models (4). As all GPCRs, the CRF1R consists of an extracellular amino-terminal domain (N-domain) and seven α-helical transmembrane domains (TM1-TM7), which are connected extracellularly with three loops (EL1-EL3). The CRF and its related peptides, such as sauvagine, bind to the extracellular regions of CRF1R and activate the receptor (5,6). In contrast, small-molecule nonpeptide antagonists have been proposed to interact with the TMs of CRF1R and allosterically antagonize peptide-agonist binding and receptor activation (7). Recently our laboratory has shown that, similar to other GPCRs, the TMs of CRF1R fold such as to form a water-accessible crevice (binding-site crevice) within the plasma membrane (8). Amino acids of TMs that contact non-peptide CRF analogues should be on the surface of the binding-site crevice of CRF1R. Moreover, TM residues in family A GPCRs have been shown to participate in networks of interactions that play important role in receptor activation (9). However the molecular mechanisms underlying CRF1R activation and its antagonism by non-peptide molecules are still elusive. The present study aims to 1) obtain structural information for the TMs of CRF1R, starting from TM3, 2) determine structural changes associated with receptor activation, 3) determine TM residues that interact with non-peptide CRF antagonists and 4) elucidate the molecular mechanisms underlying antagonism of receptor activation by non-peptide CRF antagonists. The TM3 has been selected because its tilted orientation in other GPCRs, relative to membrane, allows its residues to establish key interactions with ligands, other TMs and with G-proteins (10). To obtain structural information for the TM3 of CRF1R, we identified its residues that are located on the surface of the binding-site crevice of an inactive ligand-free state (apo-state) of the full-length receptor. To achieve this we mutated, one at a time, the TM3 residues to Cys IX (engineered Cys) and determined their accessibilities by applying the Substituted Cysteine Accessibility Method (SCAM) (11,12). Among the twenty-two TM3 residues those at positions (upper half of TM3) 1893.26, 1923.29, 1933.30, 1953.32, 1963.33, 1993.36 and 2033.40 were found to be located on the surface of the binding site crevice. These results suggest that the TM3 of the apo inactive state of CRF1R is positioned such that the upper half of this helix participates in the formation of a large binding site crevice, whereas the other half is tightly packed with the other TMs. These results are in full agreement with those obtained from a crystallization study of a Nterminally truncated inactive non-peptide ligand-bound state of CRF1R which has been published very recently. Among TM3 residues, those at positions 2033.40 and 2103.47 play crucial role in receptor activation. Mutation of G2103.47 to Cys (G2103.47C) largely decreased the high binding affinity of sauvagine, as well as, its potency to stimulate the accumulation of cAMP in cells expressing the receptor. According to our molecular model, which was constructed based on the crystal structures of the inactive glucagon receptor (GCGR) and CRF1R, the G2103.47 is located one helical turn below M2063.43. The M2063.43 forms a hydrogen bond with N2835.54 of TM5 (13,14). Mutation of G2103.47 to Cys adds a side chain at position 2103.47 which could form an additional hydrogen bond with the side chain of N2835.54 that further strengthens the TM3-TM5 interface, thus stabilizing the inactive conformation of receptor. These results suggest that movements of TM3 and TM5 that take place during activation of family A GPCRs (15), might also occur in class B receptors and are hampered by strengthening the TM3-TM5 interactions. This suggestion is further supported by the fact that mutation of G2103.47 to Ala (G2103.47A) did not largely decrease the binding affinity and potency of sauvagine. In contrast to Cys, Ala at 2103.47 position of CRF1R did not form a hydrogen bond with N2835.54, and thus having a much smaller impact on receptor activation. As G2103.47, F2033.40 in TM3 plays an important role in receptor activation. Mutation of F2033.40 to Cys (F2033.40C) largely decreased the ability of receptor to adopt its active state and to bind sauvagine with high affinity. Interestingly, addition of a Lys-like chain to F2033.40C after its reaction with the positively charged reagent, MTSEA, restored sauvagine binding to normal (high affinity) levels. Similarly, addition of a Lys side chain at position 2033.40 by mutating F2033.40 to Lys (F2033.40K) did not significantly alter the high binding affinity of sauvagine. As F2033.40K substitution, mutation of F2033.40 to Trp (F2033.40 W) or Ile (F2033.40 I) did not decrease the high affinity binding and potency of sauvagine. In marked contrast, removing the X hydrophobic side chain at position 2033.40 by mutating F2033.40 to Ala (F2033.40 A) reduced the binding affinity and potency of sauvagine. Based on these results and comparing the inactive crystal structure of CRF1R with the inactive and active crystal structures of the family A GPCR, β2 adrenergic receptor (β2AR), we propose that F2033.40 in TM3 and L3236.44 in TM6 of CRF1R, form an aromatic-aliphatic interaction in the inactive state of receptor. As in β2AR, activation of CRF1R is associated with a repositioning of F2033.40 relative to L3236.44 and towards a nearby aromatic amino acid, which possibly interacts with F2033.40, Trp (F2033.40 W) or Ile (F2033.40 I) but not with Ala (F2033.40 A). Such an aromatic-aromatic interaction most likely stabilizes the high affinity binding active state of CRF1R and it is supported by that F2033.40K mutation did not alter sauvagine binding. Lys (such as K2033.40) has been shown to participate in cation-pi interactions with aromatic residues (16,17). Despite the fact that the hydrophilic substitution F2033.40K did not alter the ability of CRF1R to adopt its active state, this modification abolished the binding of the hydrophobic nonpeptide antagonist, antalarmin to receptor. Similarly, F2033.40A mutation abolished antalarmin binding to CRF1R. In marked contrast, the hydrophobic substitutions F2033.40W and F2033.40I did not decrease the binding and antagonistic properties of antalarmin. These results suggest that F2033.40 interacts with non-peptide CRF antagonists, as also observed in the crystal structure of CRF1R in complex with the non-peptide antagonist CP-376395 (13). Interaction of non-peptide CRF antagonists with TMs of CRF1R and intercalation between them most likely hampers receptor activation-associated movements of these regions. Conclusively the present study provided structural information for the CRF1R and elucidated the molecular mechanisms underlying receptor activation and its antagonism by small non-peptide molecules. In parallel this study designed, synthesized and pharmacologically evaluated several non-peptide CRF analogues. Among the 50 compounds tested, four showed promising binding affinities for CRF1R and were selected as lead compounds for the design of novel molecules. All the results of the present study will put the basis for the design of novel non-peptide CRF1R-selective antagonists with improved pharmacodynamic and pharmacokinetic properties that will enrich the pharmaceutical armoire against CRF1R-related disorders such as depression and anxiety.
Language Greek
Subject Corticotropin releasing factor
Εκλυτικός παράγον της κορτικοτροπίνης
Issue date 2016-12-13
Collection   School/Department--School of Medicine--Department of Medicine--Doctoral theses
  Type of Work--Doctoral theses
Views 468

Digital Documents
No preview available

Download document
View document
Views : 9