Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where George Liapakis is active.

Publication


Featured researches published by George Liapakis.


Methods in Enzymology | 2002

Use of the substituted cysteine accessibility method to study the structure and function of G protein-coupled receptors.

Jonathan A. Javitch; Lei Shi; George Liapakis

Publisher Summary Cysteine substitution and covalent modification have been used to study structure–function relationships and the dynamics of protein function in a variety of membrane proteins. Charged, hydrophilic, sulfhydryl reagents have been used to probe systematically the accessibility of substituted cysteines in putative transmembrane segments of a number of proteins. This approach, the substituted cysteine accessibility method (SCAM), has been used to map channel-lining residues in the nicotinic acetylcholine receptor, the GABA receptor, the cystic fibrosis transmembrane conductance regulator, the UhpT transporter, and potassium channels, among others. This chapter uses this approach to map the surface of the binding-site crevice in the dopamine D2 receptor, a member of the G protein-coupled receptor (GPCR) superfamily. SCAM provides an approach to map systematically residues on the water-accessible surface of a protein. These residues are identified by substituting them with cysteine and assessing them for the reaction of charged, hydrophilic, sulfhydryl reagents with the engineered cysteines. Consecutive residues in putative transmembrane segments are mutated to cysteine, one at a time, and the mutant proteins are expressed in heterologous cells.


Molecular Pharmacology | 2009

Alanine Scanning Mutagenesis of the Second Extracellular Loop of Type 1 Corticotropin-Releasing Factor Receptor Revealed Residues Critical for Peptide Binding

Kostas Gkountelias; Theodoros Tselios; Maria Venihaki; George Deraos; Iakovos Lazaridis; Olga Rassouli; Achille Gravanis; George Liapakis

Upon binding of the corticotropin-releasing factor (CRF) analog sauvagine to the type 1 CRF receptor (CRF1), the amino-terminal portion of the peptide has been shown to lie near Lys257 in the receptors second extracellular loop (EL2). To test the hypothesis that EL2 residues play a role in the binding of sauvagine to CRF1 we carried out an alanine-scanning mutagenesis study to determine the functional role of EL2 residues (Leu251 to Val266). Only the W259A, F260A, and W259A/F260A mutations reduced the binding affinity and potency of sauvagine. In contrast, these mutations did not seem to significantly alter the overall receptor conformation, in that they left unchanged the affinities of the ligands astressin and antalarmin that have been suggested to bind to different regions of CRF1. The W259A, F260A, and W259A/F260A mutations also decreased the affinity of the endogenous ligand, CRF, implying that these residues may play a common important role in the binding of different peptides belonging to CRF family. Parallel amino acid deletions of the two peptides produced ligands with various affinities for wild-type CRF1 compared with the W259A, F260A, and W259A/F260A mutants, supporting the interaction between the amino-terminal residues 8 to 10 of sauvagine and the corresponding region in CRF with EL2 of CRF1. This is the first time that a specific region of CRF1 has been implicated in detailed interactions between the receptor and the amino-terminal portion of peptides belonging to the CRF family.


Current Pharmaceutical Design | 2012

The G-protein Coupled Receptor Family: Actors with Many Faces

George Liapakis; Arnau Cordomí; Leonardo Pardo

G-protein coupled receptors (GPCRs) comprise the largest family of proteins in our body, which have many important physiological functions and are implicated in the pathophysiology of many serious diseases. GPCRs therefore are significant targets in pharmaceutical research. GPCRs share the common architecture of seven plasma membrane-spanning segments connected to each other with three extracellular and three intracellular loops. In addition, GPCRs contain an extracellular N-terminal region and an intracellular C-terminal tail. GPCRs could stimulate different intracellular G-proteins (internal stimuli) and signaling pathways after their interaction with different ligands (external stimuli). The exceptional functional plasticity of GPCRs could be attributed to their inherent dynamic nature to adopt different active conformations, which are stabilized differentially by different stimuli as well as by several mutations. This review describes the structural changes of GPCRs associated with their activation. Understanding the dynamic nature of GPCRs could potentially contribute in the development of future structure-based approaches to design new receptor-specific, signaling-selective ligands, which will enrich the pharmaceutical armamentarium against various diseases.


Current Medicinal Chemistry | 2011

Members of CRF Family and their Receptors: From Past to Future

George Liapakis; Maria Venihaki; Andrew N. Margioris; D. Grigoriadis; K. Gkountelias

Corticotropin releasing factor (CRF), originally isolated from the mammalian hypothalamus, is a 41 amino acid peptide that plays an important physiological role and is implicated in the pathophysiology of various diseases. In addition to CRF and its related peptides, a large number of small non-peptide CRF analogs have been recently synthesized, some currently in clinical trials having considerable therapeutic potential in the treatment of CRF-related illnesses. CRF and its related peptides exert their multiple actions by interacting with two types of plasma membrane G-protein coupled CRF receptors, the type 1 (CRF(1)) and type 2 (CRF(2)). These receptors, like all GPCRs consist of an amino-terminal extracellular region, a carboxyl-terminal intracellular tail and seven, membrane-spanning segments, connected by alternating intracellular and extracellular loops. This review describes the functional role of CRF receptors and their ligands emphasizing the structural elements that are important for their function and could potentially contribute in the development of future target-based approaches to design new CRF-related drugs which will enrich the pharmaceutical armoire against serious diseases.


Journal of Biological Chemistry | 2012

Mapping Structural Determinants within Third Intracellular Loop That Direct Signaling Specificity of Type 1 Corticotropin-releasing Hormone Receptor

Anu Punn; Jing Chen; Maria Delidaki; Jiyou Tang; George Liapakis; Hendrik Lehnert; Michael A. Levine; Dimitris K. Grammatopoulos

Background: The CRH-R1 is key for mammalian adaptation to stress; however, the structural determinants of signal transduction are unknown. Results: Amino acids identified within CRH-R1 IC3 appear crucial for G protein-dependent cAMP and ERK1/2 signaling. Conclusion: These microdomains provide a tight control of CRH-R1 differential coupling to distinct G proteins and downstream effectors. Significance: This might prevent hyperstimulation of stress-induced responses. The type 1 corticotropin-releasing hormone receptor (CRH-R1) influences biological responses important for adaptation to stressful stimuli, through activation of multiple downstream effectors. The structural motifs within CRH-R1 that mediate G protein activation and signaling selectivity are unknown. The aim of this study was to gain insights about important structural determinants within the third intracellular loop (IC3) of the human CRH-R1α important for cAMP and ERK1/2 pathways activation and selectivity. We investigated the role of the juxtamembrane regions of IC3 by mutating amino acid cassettes or specific residues to alanine. Although simultaneous tandem alanine mutations of both juxtamembrane regions Arg292-Met295 and Lys311-Lys314 reduced ligand binding and impaired signaling, all other mutant receptors retained high affinity binding, indistinguishable from wild-type receptor. Agonist-activated receptors with tandem mutations at the proximal or distal terminal segments enhanced activation of adenylyl cyclase by 50–75% and diminished activation of inositol trisphosphate and ERK1/2 by 60–80%. Single Ala mutations identified Arg292, Lys297, Arg310, Lys311, and Lys314 as important residues for the enhanced activation of adenylyl cyclase, partly due to reduced inhibition of adenylyl cyclase activity by pertussis toxin-sensitive G proteins. In contrast, mutation of Arg299 reduced receptor signaling activity and cAMP response. Basic as well as aliphatic amino acids within both juxtamembrane regions were identified as important for ERK1/2 phosphorylation through activation of pertussis toxin-sensitive G proteins as well as Gq proteins. These data uncovered unexpected roles for key amino acids within the highly conserved hydrophobic N- and C-terminal microdomains of IC3 in the coordination of CRH-R1 signaling activity.


Journal of Pharmacology and Experimental Therapeutics | 2011

Evaluation of a Stable Gonadotropin-Releasing Hormone Analog in Mice for the Treatment of Endocrine Disorders and Prostate Cancer

Theodora Katsila; Evangelos Balafas; George Liapakis; Patrizia Limonta; Marina Montagnani Marelli; Konstantinos Gkountelias; Theodore Tselios; Nikolaos Kostomitsopoulos; John Matsoukas; Constantin Tamvakopoulos

Gonadotropin-releasing hormone (GnRH) receptor agonists have wide clinical applications including the treatment of prostate cancer and endocrine disorders. However, such agonists are characterized by poor pharmacokinetic properties, often requiring repeated administration or special formulations. Therefore, the development of novel peptide analogs with enhanced in vivo stability could potentially provide therapeutic alternatives. The pharmacological evaluation of a bioactive peptide [Des-Gly10,Tyr5(OMe),d-Leu6,Aze-NHEt9]GnRH, analog 1, is presented herein and compared with leuprolide. Peptide stability was evaluated using mouse kidney membrane preparations, followed by a liquid chromatography-tandem mass spectrometry-based approach that afforded identification and quantification of its major metabolites. The analog was significantly more stable in vitro in comparison with leuprolide. In vitro and in vivo stability results correlated well, encouraging us to develop a clinically relevant pharmacokinetic mouse model, which facilitated efficacy measurements using testosterone as a biomarker. Analog 1, an agonist of the GnRH receptor with a binding affinity in the nanomolar range, caused testosterone release in mice that was acutely dose-dependent, an effect blocked by the GnRH receptor antagonist cetrorelix. Repeated dosing studies in mice demonstrated that analog 1 was well tolerated and had potency similar to that of leuprolide, based on plasma and testis testosterone reduction and histopathological findings. Analog 1 also shared with leuprolide similar significant antiproliferative activity on androgen-dependent prostate cancer (LNCaP) cells. On the basis of pharmacokinetic advantages, we expect that analog 1 or analogs based on this new design will be therapeutically advantageous for the treatment of cancer and endocrine disorders.


Current Medicinal Chemistry | 2015

Leveraging NMR and X-ray Data of the Free Ligands to Build Better Drugs Targeting Angiotensin II Type 1 G-Protein Coupled Receptor.

Tahsin F. Kellici; Dimitrios Ntountaniotis; Eftichia Kritsi; Maria Zervou; Panagiotis Zoumpoulakis; Constantinos Potamitis; Serdar Durdagi; Ramin Ekhteiari Salmas; Gizem Ergun; Ebru Gokdemir; Maria Halabalaki; Ioannis P. Gerothanassis; George Liapakis; Andreas G. Tzakos; Thomas Mavromoustakos

The angiotensin II type 1 receptor (AT1R) has been recently crystallized. A new era has emerged for the structure-based rational drug design and the synthesis of novel AT1R antagonists. In this critical review, the X-ray crystallographic data of commercially available AT1R antagonists in free form are analyzed and compared with the conformational analysis results obtained using a combination of NMR spectroscopy and Molecular Modeling. The same AT1R antagonists are docked and compared in terms of their interactions in their binding site using homology models and the crystallized AT1R receptor. Various aspects derived from these comparisons regarding rational drug design are outlined.


Bioconjugate Chemistry | 2014

GnRH-Gemcitabine conjugates for the treatment of androgen-independent prostate cancer: pharmacokinetic enhancements combined with targeted drug delivery.

Theodoros Karampelas; Orestis Argyros; Nisar Sayyad; Katerina Spyridaki; Charalampos Pappas; Kevin Morgan; George Kolios; Robert P. Millar; George Liapakis; Andreas G. Tzakos; Demosthenes Fokas; Constantin Tamvakopoulos

Gemcitabine, a drug with established efficacy against a number of solid tumors, has therapeutic limitations due to its rapid metabolic inactivation. The aim of this study was the development of an innovative strategy to produce a metabolically stable analogue of gemcitabine that could also be selectively delivered to prostate cancer (CaP) cells based on cell surface expression of the Gonadotropin Releasing Hormone-Receptor (GnRH-R). The synthesis and evaluation of conjugated molecules, consisting of gemcitabine linked to a GnRH agonist, is presented along with results in androgen-independent prostate cancer models. NMR and ligand binding assays were employed to verify conservation of microenvironments responsible for binding of novel GnRH-gemcitabine conjugates to the GnRH-R. In vitro cytotoxicity, cellular uptake, and metabolite formation of the conjugates were examined in CaP cell lines. Selected conjugates were efficacious in the in vitro assays with one of them, namely, GSG, displaying high antiproliferative activity in CaP cell lines along with significant metabolic and pharmacokinetic advantages in comparison to gemcitabine. Finally, treatment of GnRH-R positive xenografted mice with GSG showed a significant advantage in tumor growth inhibition when compared to gemcitabine.


Journal of Biological Chemistry | 2014

Structural-Functional Analysis of the Third Transmembrane Domain of the Corticotropin-releasing Factor Type 1 Receptor ROLE IN ACTIVATION AND ALLOSTERIC ANTAGONISM

Katerina Spyridaki; Minos-Timotheos Matsoukas; Arnau Cordomí; Kostas Gkountelias; Maria Papadokostaki; Thomas Mavromoustakos; Diomedes E. Logothetis; Andrew N. Margioris; Leonardo Pardo; George Liapakis

Background: The molecular mechanisms underlying activation of CRF1 receptor (CRF1R) were elusive. Results: We determined specific residues in the transmembrane domains (TMs) of CRF1R that are critical for receptor activation. Conclusion: A possible “transmission switch” involving TM interactions is important for CRF1R activation. Significance: This knowledge may aid in the development of nonpeptide CRF1R antagonists for use in stress-related disorders. The corticotropin-releasing factor (CRF) type 1 receptor (CRF1R) for the 41-amino acid peptide CRF is a class B G protein-coupled receptor, which plays a key role in the response of our body to stressful stimuli and the maintenance of homeostasis by regulating neural and endocrine functions. CRF and related peptides, such as sauvagine, bind to the extracellular regions of CRF1R and activate the receptor. In contrast, small nonpeptide antagonists, which are effective against stress-related disorders, such as depression and anxiety, have been proposed to interact with the helical transmembrane domains (TMs) of CRF1R and allosterically antagonize peptide binding and receptor activation. Here, we aimed to elucidate the role of the third TM (TM3) in the molecular mechanisms underlying activation of CRF1R. TM3 was selected because its tilted orientation, relative to the membrane, allows its residues to establish key interactions with ligands, other TM helices, and the G protein. Using a combination of pharmacological, biochemical, and computational approaches, we found that Phe-2033.40 and Gly-2103.47 in TM3 play an important role in receptor activation. Our experimental findings also suggest that Phe-2033.40 interacts with nonpeptide antagonists.


PLOS ONE | 2014

Corticotrophin-Releasing Factor (CRF) and the Urocortins Are Potent Regulators of the Inflammatory Phenotype of Human and Mouse White Adipocytes and the Differentiation of Mouse 3T3L1 Pre-Adipocytes

Eirini Dermitzaki; George Liapakis; Ariadne Androulidaki; Maria Venihaki; John Melissas; Christos Tsatsanis; Andrew N. Margioris

Chronic activation of innate immunity takes place in obesity and initiated by the hypertrophic adipocytes which obtain a pro-inflammatory phenotype. The corticotrophin-releasing factor (CRF) family of neuropeptides and their receptors (CRF1 and CRF2) affect stress response and innate immunity. Adipose tissue expresses a complete CRF system. The aim of this study was to examine the role of CRF neuropeptides in the immune phenotype of adipocytes assessed by their expression of the toll-like receptor-4 (TLR4), the production of inflammatory cytokines IL-6, TNF-α and IL-1β, chemokines IL-8, monocyte attractant protein-1 (MCP-1) and of the adipokines adiponectin, resistin and leptin. Our data are as follows: (a) CRF, UCN2 and UCN3 are expressed in human white adipocytes as well as CRFR1a, CRFR2a and CRFR2b but not CRFR2c. 3T3L1 pre-adipocytes and differentiated adipocytes expressed both CRF1 and CRF2 receptors and UCN3, while UCN2 was detected only in differentiated adipocytes. CRF2 was up-regulated in mouse mature adipocytes. (b) CRF1 agonists suppressed media- and LPS-induced pre-adipocyte differentiation while CRF2 receptor agonists had no effect. (c) In mouse pre-adipocytes, CRF2 agonists suppressed TLR4 expression and the production of IL-6, CXCL1 and adiponectin while CRF1 agonists had no effect. (d) In mature mouse adipocytes LPS induced IL-6 and CXCL1 production and suppressed leptin. (e) In human visceral adipocytes LPS induced IL-6, TNF-α, IL-8, MCP-1 and leptin production and suppressed adiponectin and resistin. (f) In mouse mature adipocytes CRF1 and CRF2 agonists suppressed basal and LPS-induced production of inflammatory cytokines, TLR4 expression and adiponectin production, while in human visceral adipocytes CRF and UCN1 suppressed basal and LPS-induced IL-6, TNF-α, IL-8 and MCP-1 production. In conclusion, the effects of the activation of CRF1 and CRF2 may be significant in ameliorating the pro-inflammatory activity of adipocytes in obesity.

Collaboration


Dive into the George Liapakis's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Thomas Mavromoustakos

National and Kapodistrian University of Athens

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bhimanna Kuppast

South Dakota State University

View shared research outputs
Researchain Logo
Decentralizing Knowledge