Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Front Chem ; 9: 718405, 2021.
Article in English | MEDLINE | ID: mdl-34692637

ABSTRACT

Because of the key relevance of protein-protein interactions (PPI) in diseases, the modulation of protein-protein complexes is of relevant clinical significance. The successful design of binding compounds modulating PPI requires a detailed knowledge of the involved protein-protein system at molecular level, and investigation of the structural motifs that drive the association of the proteins at the recognition interface. These elements represent hot spots of the protein binding free energy, define the complex lifetime and possible modulation strategies. Here, we review the advanced technologies used to map the PPI involved in human diseases, to investigate the structure-function features of protein complexes, and to discover effective ligands that modulate the PPI for therapeutic intervention.

2.
Eur J Med Chem ; 226: 113895, 2021 Dec 15.
Article in English | MEDLINE | ID: mdl-34624821

ABSTRACT

We report the synthesis of novel first-in-class 2-oxindole-based derivatives as dual PDK1-AurA kinase inhibitors as a novel strategy to treat Ewing sarcoma. The most potent compound 12 is suitable for progression to in vivo studies. The specific attributes of 12 included nanomolar inhibitory potency against both phosphoinositide-dependent kinase-1 (PDK1) and Aurora A (AurA) kinase, with acceptable in vitro ADME-Tox properties (cytotoxicity in 2 healthy and 14 hematological and solid cancer cell-lines; inhibition of PDE4C1, SIRT7, HDAC4, HDAC6, HDAC8, HDAC9, AurB, CYP1A2, CYP2C9, CYP2C19, CYP2D6, and hERG). X-ray crystallography and docking studies led to the identification of the key AurA and PDK1/12 interactions. Finally, in vitro drug-intake kinetics and in vivo PK appear to indicate that these compounds are attractive lead-structures for the design and synthesis of PDK1/AurA dual-target molecules to further investigate the in vivo efficacy against Ewing Sarcoma.


Subject(s)
Antineoplastic Agents/pharmacology , Aurora Kinase A/antagonists & inhibitors , Drug Development , Oxindoles/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/antagonists & inhibitors , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Aurora Kinase A/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Crystallography, X-Ray , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Molecular Docking Simulation , Molecular Structure , Oxindoles/chemical synthesis , Oxindoles/chemistry , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/metabolism , Structure-Activity Relationship
3.
Int J Mol Sci ; 20(18)2019 Sep 12.
Article in English | MEDLINE | ID: mdl-31547231

ABSTRACT

NGR-hTNF is a therapeutic agent for a solid tumor that specifically targets angiogenic tumor blood vessels, through the NGR motif. Its activity has been assessed in several clinical studies encompassing tumors of different histological types. The drug's activity is based on an improved permeabilization of newly formed tumor vasculature, which favors intratumor penetration of chemotherapeutic agents and leukocyte trafficking. This work investigated the binding and the signaling properties of the NGR-hTNF, to elucidate its mechanism of action. The crystal structure of NGR-hTNF and modeling of its interaction with TNFR suggested that the NGR region is available for binding to a specific receptor. Using 2D TR-NOESY experiments, this study confirmed that the NGR-peptides binds to a specific CD13 isoform, whose expression is restricted to tumor vasculature cells, and to some tumor cell lines. The interaction between hTNF or NGR-hTNF with immobilized TNFRs showed similar kinetic parameters, whereas the competition experiments performed on the cells expressing both TNFR and CD13 showed that NGR-hTNF had a higher binding affinity than hTNF. The analysis of the NGR-hTNF-triggered signal transduction events showed a specific impairment in the activation of pro-survival pathways (Ras, Erk and Akt), compared to hTNF. Since a signaling pattern identical to NGR-hTNF was obtained with hTNF and NGR-sequence given as distinct molecules, the inhibition observed on the survival pathways was presumably due to a direct effect of the NGR-CD13 engagement on the TNFR signaling pathway. The reduced activation of the pro survival pathways induced by NGR-hTNF correlated with the increased caspases activation and reduced cell survival. This study demonstrates that the binding of the NGR-motif to CD13 determines not only the homing of NGR-hTNF to tumor vessels, but also the increase in its antiangiogenic activity.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Neoplasms/blood supply , Oligopeptides/pharmacology , Recombinant Fusion Proteins/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Angiogenesis Inhibitors/chemistry , Cell Line, Tumor , Crystallography, X-Ray , Human Umbilical Vein Endothelial Cells , Humans , Models, Molecular , Oligopeptides/chemistry , Recombinant Fusion Proteins/chemistry , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/chemistry
4.
IUCrJ ; 6(Pt 2): 178-188, 2019 Mar 01.
Article in English | MEDLINE | ID: mdl-30867915

ABSTRACT

Recent advances in 3D electron diffraction have allowed the structure determination of several model proteins from submicrometric crystals, the unit-cell parameters and structures of which could be immediately validated by known models previously obtained by X-ray crystallography. Here, the first new protein structure determined by 3D electron diffraction data is presented: a previously unobserved polymorph of hen egg-white lysozyme. This form, with unit-cell parameters a = 31.9, b = 54.4, c = 71.8 Å, ß = 98.8°, grows as needle-shaped submicrometric crystals simply by vapor diffusion starting from previously reported crystallization conditions. Remarkably, the data were collected using a low-dose stepwise experimental setup consisting of a precession-assisted nanobeam of ∼150 nm, which has never previously been applied for solving protein structures. The crystal structure was additionally validated using X-ray synchrotron-radiation sources by both powder diffraction and single-crystal micro-diffraction. 3D electron diffraction can be used for the structural characterization of submicrometric macromolecular crystals and is able to identify novel protein polymorphs that are hardly visible in conventional X-ray diffraction experiments. Additionally, the analysis, which was performed on both nanocrystals and microcrystals from the same crystallization drop, suggests that an integrated view from 3D electron diffraction and X-ray microfocus diffraction can be applied to obtain insights into the molecular dynamics during protein crystal growth.

5.
ACS Chem Biol ; 13(8): 2082-2093, 2018 08 17.
Article in English | MEDLINE | ID: mdl-29878744

ABSTRACT

Reversibly photoswitchable fluorescent proteins (RSFPs) admirably combine the genetic encoding of fluorescence with the ability to repeatedly toggle between a bright and dark state, adding a new temporal dimension to the fluorescence signal. Accordingly, in recent years RSFPs have paved the way to novel applications in cell imaging that rely on their reversible photoswitching, including many super-resolution techniques such as F-PALM, RESOLFT, and SOFI that provide nanoscale pictures of the living matter. Yet many RSFPs have been engineered by a rational approach only to a limited extent, in the absence of clear structure-property relationships that in most cases make anecdotic the emergence of the photoswitching. We reported [ Bizzarri et al. J. Am Chem Soc. 2010 , 102 , 85 ] how the E222Q replacement is a single photoswitching mutation, since it restores the intrinsic cis-trans photoisomerization properties of the chromophore in otherwise nonswitchable Aequorea proteins of different color and mutation pattern (Q-RSFPs). We here investigate the subtle role of Q222 on the excited-state photophysics of the two simplest Q-RSFPs by a combined experimental and theoretical approach, using their nonswitchable anacestor EGFP as benchmark. Our findings link indissolubly photoswitching and Q222 presence, by a simple yet elegant scenario: largely twisted chromophore structures around the double bond (including hula-twist configurations) are uniquely stabilized by Q222 via H-bonds. Likely, these H-bonds subtly modulate the electronic properties of the chromophore, enabling the conical intersection that connects the excited cis to ground trans chromophore. Thus, Q222 belongs to a restricted family of single mutations that change dramatically the functional phenotype of a protein. The capability to distinguish quantitatively T65S/E222Q EGFP ("WildQ", wQ) from the spectrally identical EGFP by quantitative Optical Lock-In Detection (qOLID) witnesses the relevance of this mutation for cell imaging.


Subject(s)
Hydrozoa/chemistry , Luminescent Proteins/chemistry , Animals , CHO Cells , Cricetulus , Green Fluorescent Proteins/chemistry , Hydrogen Bonding , Isomerism , Light , Models, Molecular , Optical Imaging , Photochemical Processes
6.
Chem Commun (Camb) ; 53(95): 12814-12817, 2017 Nov 28.
Article in English | MEDLINE | ID: mdl-29143042

ABSTRACT

N-Acylphosphatidylethanolamine phospholipase D (NAPE-PLD) is a membrane-associated zinc enzyme that catalyzes the hydrolysis of N-acylphosphatidylethanolamines (NAPEs) into fatty acid ethanolamides (FAEs). Here, we describe the identification of the first small-molecule NAPE-PLD inhibitor, the quinazoline sulfonamide derivative 2,4-dioxo-N-[4-(4-pyridyl)phenyl]-1H-quinazoline-6-sulfonamide, ARN19874.


Subject(s)
Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Phospholipase D/antagonists & inhibitors , Quinazolines/chemical synthesis , Quinazolines/pharmacology , Small Molecule Libraries/pharmacology , Sulfonamides/chemical synthesis , Sulfonamides/pharmacology , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Humans , Models, Molecular , Molecular Structure , Phospholipase D/metabolism , Quinazolines/chemistry , Quinazolinones , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Structure-Activity Relationship , Sulfonamides/chemistry
7.
J Mater Chem B ; 5(2): 279-288, 2017 Jan 14.
Article in English | MEDLINE | ID: mdl-32263546

ABSTRACT

Hydroxyapatite bioactive complexes are being increasingly recognized as effective available means in regenerative medicine. Conventional technologies for their synthesis have drawbacks from a synthetic standpoint, mainly requiring high temperatures and multi-step processes. Here, we show that ultra-small hydroxyapatite conjugated-nanoparticles (Ha-CNPs) can be obtained at room temperature by Pulsed Laser Ablation (PLA) directly in protein solution using picosecond pulses at near infrared wavelengths. The results showed that the nanoparticle size was driven by the concentration of the protein. Using this approach, we obtained aqueous soluble and ultra-small crystalline nanoparticles of ≈3 nm diameter coated with protein molecules (surface coverage ≈ 5.5 pmol cm-2; zeta potential ≈-33.5 mV). These nanoparticles showed low cytotoxicity in vitro compared to chemically synthesized nanoparticles, and revealed proliferative and osteoinductive effects on human bone marrow mesenchymal stem cells (hMSCs). The resulting enhanced cell osteogenic differentiation suggested that our PLA-based synthetic approach might be exploited in novel applications of regenerative medicine.

8.
ACS Chem Biol ; 11(10): 2908-2914, 2016 10 21.
Article in English | MEDLINE | ID: mdl-27571266

ABSTRACT

The membrane-associated enzyme NAPE-PLD (N-acyl phosphatidylethanolamine specific-phospholipase D) generates the endogenous cannabinoid arachidonylethanolamide and other lipid signaling amides, including oleoylethanolamide and palmitoylethanolamide. These bioactive molecules play important roles in several physiological pathways including stress and pain response, appetite, and lifespan. Recently, we reported the crystal structure of human NAPE-PLD and discovered specific binding sites for the bile acid deoxycholic acid. In this study, we demonstrate that in the presence of this secondary bile acid, the stiffness of the protein measured by elastic neutron scattering increases, and NAPE-PLD is ∼7 times faster to catalyze the hydrolysis of the more unsaturated substrate N-arachidonyl-phosphatidylethanolamine, compared with N-palmitoyl-phosphatidylethanolamine. Chenodeoxycholic acid and glyco- or tauro-dihydroxy conjugates can also bind to NAPE-PLD and drive its activation. The only natural monohydroxy bile acid, lithocholic acid, shows an affinity of ∼20 µM and acts instead as a reversible inhibitor (IC50 ≈ 68 µM). Overall, these findings provide important insights into the allosteric regulation of the enzyme mediated by bile acid cofactors and reveal that NAPE-PLD responds primarily to the number and position of their hydroxyl groups.


Subject(s)
Bile Acids and Salts/metabolism , Phospholipase D/metabolism , Bile Acids and Salts/chemistry , Crystallography, X-Ray , Humans , Kinetics , Phospholipase D/chemistry , Protein Conformation , Structure-Activity Relationship , Substrate Specificity
9.
Anal Biochem ; 495: 52-9, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26686030

ABSTRACT

Nuclear magnetic resonance (NMR)-based screening has been recognized as a powerful approach for the identification and characterization of molecules interacting with pharmaceutical targets. Indeed, several NMR methods have been developed and successfully applied to many drug discovery projects. Whereas most of these approaches have targeted isolated biomolecular receptors, very few cases are reported with the screening performed in intact cells and cell extracts. Here we report the first successful application of the fluorine NMR-based assay n-FABS (n-fluorine atoms for biochemical screening) in living mammalian cells expressing the membrane protein fatty acid amide hydrolase (FAAH). This method allows the identification of both weak and potent inhibitors and the measurement of their potency in a physiological environment.


Subject(s)
Amidohydrolases/analysis , Nuclear Magnetic Resonance, Biomolecular , Amidohydrolases/metabolism , Benzamides/chemistry , Benzamides/metabolism , Carbamates/chemistry , Carbamates/metabolism , Fluorine/chemistry , HEK293 Cells , Humans , Inhibitory Concentration 50
10.
Exp Neurol ; 274(Pt B): 156-65, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26272754

ABSTRACT

Our previous studies revealed that L-type voltage-dependent Ca(2+) channels (Cav1.2 L-VDCCs) are modulated by the neural extracellular matrix backbone, polyanionic glycan hyaluronic acid. Here we used isothermal titration calorimetry and screened a set of peptides derived from the extracellular domains of Cav1.2α1 to identify putative binding sites between the channel and hyaluronic acid or another class of polyanionic glycans, such as heparin/heparan sulfates. None of the tested peptides showed detectable interaction with hyaluronic acid, but two peptides derived from the first pore-forming domain of Cav1.2α1 subunit bound to heparin. At 25 °C the binding of the peptide P7 (MGKMHKTCYN) was at ~50 µM, and that of the peptide P8 (GHGRQCQNGTVCKPGWDGPKHG) was at ~21 µM. The Cav1.2α1 first pore forming segment that contained both peptides maintained a high affinity for heparin (~23 µM), integrating their enthalpic and entropic binding contributions. Interaction between heparin and recombinant as well as native full-length neuronal Cav1.2α1 channels was confirmed using the heparin-agarose pull down assay. Whole cell patch clamp recordings in HEK293 cells transfected with neuronal Cav1.2 channels revealed that enzymatic digestion of highly sulfated heparan sulfates with heparinase 1 affects neither voltage-dependence of channel activation nor the level of steady state inactivation, but did speed up channel inactivation. Treatment of hippocampal cultures with heparinase 1 reduced the firing rate and led to appearance of long-lasting bursts in the same manner as treatment with the inhibitor of L-VDCC diltiazem. Thus, heparan sulfate proteoglycans may bind to and regulate L-VDCC inactivation and network activity.


Subject(s)
Calcium Channels, L-Type/metabolism , Heparin/pharmacology , Heparitin Sulfate/metabolism , Neurons/metabolism , Action Potentials/drug effects , Action Potentials/genetics , Animals , Binding Sites/drug effects , Binding Sites/genetics , Biophysical Phenomena/drug effects , Biophysical Phenomena/genetics , CHO Cells , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/chemistry , Calcium Channels, L-Type/genetics , Cricetulus , Diltiazem/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , HEK293 Cells , Heparin Lyase/pharmacology , Hippocampus/cytology , Hippocampus/drug effects , Humans , Microsomes/drug effects , Microsomes/metabolism , Models, Molecular , Nerve Net/drug effects , Neurons/drug effects , Protein Binding/drug effects , Time Factors , Valine/analogs & derivatives , Valine/pharmacology
11.
ACS Chem Biol ; 10(9): 2057-2064, 2015 Sep 18.
Article in English | MEDLINE | ID: mdl-26102511

ABSTRACT

N-Acylethanolamine acid amidase (NAAA) is a lysosomal cysteine hydrolase involved in the degradation of saturated and monounsaturated fatty acid ethanolamides (FAEs), a family of endogenous lipid signaling molecules that includes oleoylethanolamide (OEA) and palmitoylethanolamide (PEA). Among the reported NAAA inhibitors, α-amino-ß-lactone (3-aminooxetan-2-one) derivatives have been shown to prevent FAE hydrolysis in innate-immune and neural cells and to reduce reactions to inflammatory stimuli. Recently, we disclosed two potent and selective NAAA inhibitors, the compounds ARN077 (5-phenylpentyl-N-[(2S,3R)-2-methyl-4-oxo-oxetan-3-yl]carbamate) and ARN726 (4-cyclohexylbutyl-N-[(S)-2-oxoazetidin-3-yl]carbamate). The former is active in vivo by topical administration in rodent models of hyperalgesia and allodynia, while the latter exerts systemic anti-inflammatory effects in mouse models of lung inflammation. In the present study, we designed and validated a derivative of ARN726 as the first activity-based protein profiling (ABPP) probe for the in vivo detection of NAAA. The newly synthesized molecule 1 is an effective in vitro and in vivo click-chemistry activity based probe (ABP), which is able to capture the catalytically active form of NAAA in Human Embryonic Kidney 293 (HEK293) cells overexpressing human NAAA as well as in rat lung tissue. Competitive ABPP with 1 confirmed that ARN726 and ARN077 inhibit NAAA in vitro and in vivo. Compound 1 is a useful new tool to identify activated NAAA both in vitro and in vivo and to investigate the physiological and pathological roles of this enzyme.


Subject(s)
Amidohydrolases/metabolism , Enzyme Assays/methods , Molecular Probes/metabolism , Amidohydrolases/analysis , Amidohydrolases/antagonists & inhibitors , Animals , HEK293 Cells , Humans , Lung/enzymology , Male , Molecular Probes/chemistry , Rats, Sprague-Dawley
12.
Structure ; 23(3): 598-604, 2015 Mar 03.
Article in English | MEDLINE | ID: mdl-25684574

ABSTRACT

The fatty acid ethanolamides (FAEs) are lipid mediators present in all organisms and involved in highly conserved biological functions, such as innate immunity, energy balance, and stress control. They are produced from membrane N-acylphosphatidylethanolamines (NAPEs) and include agonists for G protein-coupled receptors (e.g., cannabinoid receptors) and nuclear receptors (e.g., PPAR-α). Here, we report the crystal structure of human NAPE-hydrolyzing phospholipase D (NAPE-PLD) at 2.65 Å resolution, a membrane enzyme that catalyzes FAE formation in mammals. NAPE-PLD forms homodimers partly separated by an internal ∼ 9-Å-wide channel and uniquely adapted to associate with phospholipids. A hydrophobic cavity provides an entryway for NAPE into the active site, where a binuclear Zn(2+) center orchestrates its hydrolysis. Bile acids bind with high affinity to selective pockets in this cavity, enhancing dimer assembly and enabling catalysis. These elements offer multiple targets for the design of small-molecule NAPE-PLD modulators with potential applications in inflammation and metabolic disorders.


Subject(s)
Phospholipase D/chemistry , Bile Acids and Salts/physiology , Catalytic Domain , Crystallography, X-Ray , Humans , Hydrogen Bonding , Hydrolysis , Models, Molecular , Phosphatidylethanolamines/biosynthesis , Phospholipase D/physiology , Protein Interaction Domains and Motifs , Protein Structure, Quaternary
13.
ChemMedChem ; 9(2): 286-9, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24339446

ABSTRACT

The possibility of measuring the action of inhibitors of specific enzymatic reactions in intact cells, cell lysates or membrane preparations represents a major advance in the lead discovery process. Despite the relevance of assaying in physiological conditions, only a small number of biophysical techniques, often requiring complex set-up, are applicable to these sample types. Here, we demonstrate the first application of n-fluorine atoms for biochemical screening (n-FABS), a homogeneous and versatile assay based on (19) F NMR spectroscopy, to the detection of high- and low-affinity inhibitors of a membrane enzyme in cell extracts and determination of their IC50 values. Our approach can allow the discovery of novel binding fragments against targets known to be difficult to purify or where membrane-association is required for activity. These results pave the way for future applications of the methodology to these relevant and complex biological systems.


Subject(s)
Cell Membrane/enzymology , Drug Discovery/methods , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/metabolism , Cell Membrane/drug effects , Fluorine/analysis , Humans , Membrane Proteins/chemistry , Nuclear Magnetic Resonance, Biomolecular/methods
14.
Chembiochem ; 14(13): 1611-9, 2013 Sep 02.
Article in English | MEDLINE | ID: mdl-23918626

ABSTRACT

Despite the recognized importance of membrane proteins as pharmaceutical targets, the reliable identification of fragment hits that are able to bind these proteins is still a major challenge. Among different ¹9F NMR spectroscopic methods, n-fluorine atoms for biochemical screening (n-FABS) is a highly sensitive technique that has been used efficiently for fragment screening, but its application for membrane enzymes has not been reported yet. Herein, we present the first successful application of n-FABS to the discovery of novel fragment hits, targeting the membrane-bound enzyme fatty acid amide hydrolase (FAAH), using a library of fluorinated fragments generated based on the different local environment of fluorine concept. The use of the recombinant fusion protein MBP-FAAH and the design of compound 11 as a suitable novel fluorinated substrate analogue allowed n-FABS screening to be efficiently performed using a very small amount of enzyme. Notably, we have identified 19 novel fragment hits that inhibit FAAH with a median effective concentration (IC50) in the low mM-µM range. To the best of our knowledge, these results represent the first application of a ¹9F NMR fragment-based functional assay to a membrane protein.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Amidohydrolases/metabolism , Drug Evaluation, Preclinical/methods , Enzyme Inhibitors/analysis , Enzyme Inhibitors/pharmacology , Nuclear Magnetic Resonance, Biomolecular , Animals , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Fluorine/chemistry , Halogenation , Inhibitory Concentration 50 , Molecular Structure , Rats , Structure-Activity Relationship , Substrate Specificity
15.
J Am Chem Soc ; 135(1): 22-5, 2013 Jan 09.
Article in English | MEDLINE | ID: mdl-23240907

ABSTRACT

In addition to inhibiting the cyclooxygenase (COX)-mediated biosynthesis of prostanoids, various widely used nonsteroidal anti-inflammatory drugs (NSAIDs) enhance endocannabinoid signaling by blocking the anandamide-degrading membrane enzyme fatty acid amide hydrolase (FAAH). The X-ray structure of FAAH in complex with the NSAID carprofen, along with site-directed mutagenesis, enzyme activity assays, and NMR analysis, has revealed the molecular details of this interaction, providing information that may guide the design of dual FAAH-COX inhibitors with superior analgesic efficacy.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Carbazoles/pharmacology , Amidohydrolases/metabolism , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Binding Sites/drug effects , Carbazoles/chemistry , Dose-Response Relationship, Drug , Models, Molecular , Molecular Structure , Structure-Activity Relationship
16.
ACS Med Chem Lett ; 3(5): 422-6, 2012 May 10.
Article in English | MEDLINE | ID: mdl-24900487

ABSTRACT

The cysteine amidase N-acylethanolamine acid amidase (NAAA) is a member of the N-terminal nucleophile class of enzymes and a potential target for anti-inflammatory drugs. We investigated the mechanism of inhibition of human NAAA by substituted ß-lactones. We characterized pharmacologically a representative member of this class, ARN077, and showed, using high-resolution liquid chromatography-tandem mass spectrometry, that this compound forms a thioester bond with the N-terminal catalytic cysteine in human NAAA.

17.
Nat Neurosci ; 15(1): 64-9, 2011 Nov 20.
Article in English | MEDLINE | ID: mdl-22101642

ABSTRACT

The endocannabinoid anandamide is removed from the synaptic space by a selective transport system, expressed in neurons and astrocytes, that remains molecularly uncharacterized. Here we describe a partly cytosolic variant of the intracellular anandamide-degrading enzyme fatty acid amide hydrolase-1 (FAAH-1), termed FAAH-like anandamide transporter (FLAT), that lacked amidase activity but bound anandamide with low micromolar affinity and facilitated its translocation into cells. Known anandamide transport inhibitors, such as AM404 and OMDM-1, blocked these effects. We also identified a competitive antagonist of the interaction of anandamide with FLAT, the phthalazine derivative ARN272, that prevented anandamide internalization in vitro, interrupted anandamide deactivation in vivo and exerted profound analgesic effects in rodent models of nociceptive and inflammatory pain, which were mediated by CB(1) cannabinoid receptors. The results identify FLAT as a critical molecular component of anandamide transport in neural cells and a potential target for therapeutic drugs.


Subject(s)
Amidohydrolases/metabolism , Arachidonic Acids/metabolism , Neurons/metabolism , Polyunsaturated Alkamides/metabolism , Amidohydrolases/genetics , Animals , Arachidonic Acids/pharmacology , Benzyl Compounds/pharmacology , Endocannabinoids , Neurons/drug effects , Protein Transport/drug effects , Protein Transport/physiology , Rats
18.
J Am Chem Soc ; 132(49): 17570-7, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21082835

ABSTRACT

Conformational changes modulate macromolecular function by promoting the specific binding of ligands (such as in antigen recognition) or the stabilization of transition states in enzymatic reactions. However, quantitative characterization of the energetics underlying dynamic structural interconversions is still challenging and lacks a unified method. Here, we introduce a novel in silico approach based on the combined use of essential dynamics sampling and nonequilibrium free-energy calculations to obtain quantitative data on conformational energy landscapes. This technique allows the unbiased investigation of highly complex rearrangements, and does not require the crucial definition of user-defined collective variables. We show that free-energy values derived from profiles connecting the unliganded and ligand-bound X-ray structures of a bacterial nucleoside hydrolase match the experimental binding constant. This approach also provides first evidence for a rate-limiting character of the conformational transition in this enzyme, and an unexpected role of the protonation state of a single residue in regulating substrate binding and product release.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Escherichia coli/enzymology , Imino Furanoses/metabolism , N-Glycosyl Hydrolases/chemistry , N-Glycosyl Hydrolases/metabolism , Phenylenediamines/metabolism , Crystallography, X-Ray , Escherichia coli/chemistry , Escherichia coli/metabolism , Imino Furanoses/chemistry , Ligands , Molecular Dynamics Simulation , Phenylenediamines/chemistry , Protein Binding , Protein Conformation , Thermodynamics
19.
BMC Struct Biol ; 10: 14, 2010 Jun 08.
Article in English | MEDLINE | ID: mdl-20529317

ABSTRACT

BACKGROUND: Pyrimidine-preferring N-ribohydrolases (CU-NHs) are a class of Ca2+-dependent enzymes that catalyze the hydrolytic cleavage of the N-glycosidic bond in pyrimidine nucleosides. With the exception of few selected organisms, their physiological relevance in prokaryotes and eukaryotes is yet under investigation. RESULTS: Here, we report the first crystal structure of a CU-NH bound to a competitive inhibitor, the complex between the Escherichia coli enzyme RihA bound to 3, 4-diaminophenyl-iminoribitol (DAPIR) to a resolution of 2.1 A. The ligand can bind at the active site in two distinct orientations, and the stabilization of two flexible active site regions is pivotal to establish the interactions required for substrate discrimination and catalysis. CONCLUSIONS: A comparison with the product-bound RihA structure allows a rationalization of the structural rearrangements required for an enzymatic catalytic cycle, highlighting a substrate-assisted cooperative motion, and suggesting a yet overlooked role of the conserved His82 residue in modulating product release. Differences in the structural features of the active sites in the two homologous CU-NHs RihA and RihB from E. coli provide a rationale for their fine differences in substrate specificity. These new findings hint at a possible role of CU-NHs in the breakdown of modified nucleosides derived from RNA molecules.


Subject(s)
Binding, Competitive , Catalytic Domain , Enzyme Inhibitors/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Escherichia coli/enzymology , Imino Furanoses/metabolism , N-Glycosyl Hydrolases/chemistry , N-Glycosyl Hydrolases/metabolism , Phenylenediamines/metabolism , Ribitol/analogs & derivatives , Biocatalysis , Crystallography, X-Ray , Electrons , Enzyme Inhibitors/pharmacology , Escherichia coli Proteins/antagonists & inhibitors , Imino Furanoses/pharmacology , Models, Molecular , N-Glycosyl Hydrolases/antagonists & inhibitors , Phenylenediamines/pharmacology , Pyrimidine Nucleosides/chemistry , Pyrimidine Nucleosides/metabolism , Ribitol/metabolism , Ribitol/pharmacology , Substrate Specificity
20.
Org Biomol Chem ; 6(13): 2282-94, 2008 Jul 07.
Article in English | MEDLINE | ID: mdl-18563261

ABSTRACT

The development of broad-spectrum metallo-beta-lactamase (MBL) inhibitors is challenging due to structural diversity and differences in metal utilisation by these enzymes. Analysis of structural data, followed by non-denturing mass spectrometric analyses, identified thiols proposed to inhibit representative MBLs from all three sub-classes: B1, B2 and B3. Solution analyses led to the identification of broad spectrum inhibitors, including potent inhibitors of the CphA MBL (Aeromonas hydrophila). Structural studies revealed that, as observed for other B1 and B3 MBLs, inhibition of the L1 MBL thiols involves metal chelation. Evidence is reported that this is not the case for inhibition of the CphA enzyme by some thiols; the crystal structure of the CphA-Zn-inhibitor complex reveals a binding mode in which the thiol does not interact with the zinc. The structural data enabled the design and the production of further more potent inhibitors. Overall the results suggest that the development of reasonably broad-spectrum MBL inhibitors should be possible.


Subject(s)
Drug Evaluation, Preclinical/methods , Sulfhydryl Compounds/chemistry , Sulfhydryl Compounds/pharmacology , beta-Lactamase Inhibitors , beta-Lactamases/chemistry , Bacteria/drug effects , Bacteria/enzymology , Binding Sites , Crystallography, X-Ray , Magnetic Resonance Spectroscopy , Mass Spectrometry , Models, Molecular , Molecular Conformation
SELECTION OF CITATIONS
SEARCH DETAIL
...