151
|
Long JZ, Cravatt BF. The metabolic serine hydrolases and their functions in mammalian physiology and disease. Chem Rev 2011; 111:6022-63. [PMID: 21696217 PMCID: PMC3192302 DOI: 10.1021/cr200075y] [Citation(s) in RCA: 323] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Jonathan Z. Long
- The Skaggs Institute for Chemical Biology and Department of Chemical Physiology, The Scripps Research Institute, 10550 N. Torrey Pines Rd. La Jolla, CA 92037
| | - Benjamin F. Cravatt
- The Skaggs Institute for Chemical Biology and Department of Chemical Physiology, The Scripps Research Institute, 10550 N. Torrey Pines Rd. La Jolla, CA 92037
| |
Collapse
|
152
|
Palermo G, Branduardi D, Masetti M, Lodola A, Mor M, Piomelli D, Cavalli A, De Vivo M. Covalent inhibitors of fatty acid amide hydrolase: a rationale for the activity of piperidine and piperazine aryl ureas. J Med Chem 2011; 54:6612-23. [PMID: 21830831 DOI: 10.1021/jm2004283] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Recently, covalent drugs have attracted great interest in the drug discovery community, with successful examples that have demonstrated their therapeutic effects. Here, we focus on the covalent inhibition of the fatty acid amide hydrolase (FAAH), which is a promising strategy in the treatment of pain and inflammation. Among the most recent and potent FAAH inhibitors (FAAHi), there are the cyclic piperidine and piperazine aryl ureas. FAAH hydrolyzes efficiently the amide bond of these compounds, forming a covalent enzyme-inhibitor adduct. To rationalize this experimental evidence, we performed an extensive computational analysis centered on piperidine-based PF750 (1) and piperazine-based JNJ1661010 (2), two potent lead compounds used to generate covalent inhibitors as clinical candidates. We found that FAAH induces a distortion of the amide bond of the piperidine and piperazine aryl ureas. Quantum mechanics/molecular mechanics ΔE(LUMO-HOMO) energies indicate that the observed enzyme-induced distortion of the amide bond favors the formation of a covalent FAAH-inhibitor adduct. These findings could help in the rational structure-based design of novel covalent FAAHi.
Collapse
Affiliation(s)
- Giulia Palermo
- Department of Drug Discovery and Development, Istituto Italiano di Tecnologia , Via Morego 30, 16163 Genova, Italy
| | | | | | | | | | | | | | | |
Collapse
|
153
|
Ramikie TS, Patel S. Endocannabinoid signaling in the amygdala: anatomy, synaptic signaling, behavior, and adaptations to stress. Neuroscience 2011; 204:38-52. [PMID: 21884761 DOI: 10.1016/j.neuroscience.2011.08.037] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2011] [Revised: 08/08/2011] [Accepted: 08/17/2011] [Indexed: 01/09/2023]
Abstract
The molecular constituents of endocannabinoid (eCB) signaling are abundantly expressed within the mammalian amygdaloid complex, consistent with the robust role of eCB signaling in the modulation of emotional behavior, learning, and stress-response physiology. Here, we detail the anatomical distribution of eCB signaling machinery in the amygdala and the role of this system in the modulation of excitatory and inhibitory neuroplasticity in this region. We also summarize recent findings demonstrating dynamic alternations in eCB signaling that occur in response to stress exposure, as well as known behavioral consequences of eCB-mediated modulation of amygdala function. Finally, we discuss how integrating anatomical and physiological data regarding eCB signaling in the amygdala could help elucidate common functional motifs of this system in relation to broader forebrain function.
Collapse
Affiliation(s)
- T S Ramikie
- Neuroscience Graduate Program, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | | |
Collapse
|
154
|
Otrubova K, Ezzili C, Boger DL. The discovery and development of inhibitors of fatty acid amide hydrolase (FAAH). Bioorg Med Chem Lett 2011; 21:4674-85. [PMID: 21764305 PMCID: PMC3146581 DOI: 10.1016/j.bmcl.2011.06.096] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2011] [Revised: 06/18/2011] [Accepted: 06/20/2011] [Indexed: 11/16/2022]
Abstract
A summary of the discovery and advancement of inhibitors of fatty acid amide hydrolase (FAAH) is presented.
Collapse
Affiliation(s)
- Katerina Otrubova
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey, Pines Road, La Jolla, CA 92037, USA
| | | | | |
Collapse
|
155
|
Saario SM, McKinney MK, Speers AE, Wang C, Cravatt BF. Clickable, photoreactive inhibitors to probe the active site microenvironment of fatty acid amide hydrolase(). Chem Sci 2011; 3:77-83. [PMID: 22737400 DOI: 10.1039/c1sc00336d] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Fatty acid amide hydrolase (FAAH) is an integral membrane enzyme that degrades the endocannabinoid anandamide (AEA) and several other bioactive lipid amides. The catalytic mechanism of FAAH has been largely elucidated, and structural models of the enzyme suggest that it may recruit its hydrophobic substrates directly from the lipid bilayer of the cell. Testing this hypothesis, however, requires new tools to explore FAAH-substrate interactions in native cell membranes. Here, we have addressed this problem by creating clickable, photoreactive inhibitors that probe the microenvironment surrounding the FAAH active site. We show that these probes can be used directly in cell membranes, where distinct crosslinked adducts are observed for inhibitors that are buried within versus exposed to the external environment of the FAAH active site.
Collapse
Affiliation(s)
- Susanna M Saario
- The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA, 92037; ; Tel: +858-784-8633
| | | | | | | | | |
Collapse
|
156
|
|
157
|
Tian G, Paschetto KA, Gharahdaghi F, Gordon E, Wilkins DE, Luo X, Scott CW. Mechanism of inhibition of fatty acid amide hydrolase by sulfonamide-containing benzothiazoles: long residence time derived from increased kinetic barrier and not exclusively from thermodynamic potency. Biochemistry 2011; 50:6867-78. [PMID: 21728345 DOI: 10.1021/bi200552p] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Fatty acid amide hydrolase (FAAH) has emerged as a potential target for developing analgesic, anxiolytic, antidepressant, sleep-enhancing, and anti-inflammatory drugs, and tremendous efforts have been made to discover potent and selective inhibitors of FAAH. Most known potent FAAH inhibitors described to date employ covalent mechanisms, inhibiting the enzyme either reversibly or irreversibly. Recently, a benzothiazole-based analogue (1) has been described possessing a high potency against FAAH yet lacking a structural feature previously known to interact with FAAH covalently. However, covalent inhibition of FAAH by 1 has not been fully ruled out, and the issue of reversibility has not been addressed. Confirming previous reports, 1 inhibited recombinant human FAAH (rhFAAH) with high potency with IC(50) ~2 nM. It displayed an apparently noncompetitive and irreversible inhibition, titrating rhFAAH stoichiometrically within normal assay times. The inhibition appeared to be time dependent, but the time dependence only improved potency by a small degree (from ~8 to ~2 nM). However, mass spectrometric analyses of the reaction mixture failed to reveal any cleavage product or covalent adduct and showed full recovery of the parent compound, ruling out covalent, irreversible inhibition. Dialysis revealed recovery of enzyme activity from enzyme-inhibitor complex over a prolonged time (>10 h), demonstrating that 1 is indeed a reversible, albeit slowly dissociating inhibitor of FAAH. Molecular docking indicated that the sulfonamide group of 1 could form hydrogen bonds with several residues involved in catalysis, thereby mimicking the transition state. The long residence time displayed by 1 does not appear to derive exclusively from great thermodynamic potency and is consistent with an increased kinetic energy barrier that prevents dissociation from happening quickly.
Collapse
Affiliation(s)
- Gaochao Tian
- AstraZeneca Pharmaceuticals, Wilmington, Delaware 19850, United States.
| | | | | | | | | | | | | |
Collapse
|
158
|
He HT, Marguet D. Detecting nanodomains in living cell membrane by fluorescence correlation spectroscopy. Annu Rev Phys Chem 2011; 62:417-36. [PMID: 21219145 DOI: 10.1146/annurev-physchem-032210-103402] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Cell membranes actively participate in numerous cellular functions. Inasmuch as bioactivities of cell membranes are known to depend crucially on their lateral organization, much effort has been focused on deciphering this organization on different length scales. Within this context, the concept of lipid rafts has been intensively discussed over recent years. In line with its ability to measure diffusion parameters with great precision, fluorescence correlation spectroscopy (FCS) measurements have been made in association with innovative experimental strategies to monitor modes of molecular lateral diffusion within the plasma membrane of living cells. These investigations have allowed significant progress in the characterization of the cell membrane lateral organization at the suboptical level and have provided compelling evidence for the in vivo existence of raft nanodomains. We review these FCS-based studies and the characteristic structural features of raft nanodomains. We also discuss the findings in regards to the current view of lipid rafts as a general membrane-organizing principle.
Collapse
Affiliation(s)
- Hai-Tao He
- Centre d'Immunologie de Marseille-Luminy, INSERM UMR-S 631, and Université de la Méditerranée, F-13288 Marseille cedex 09, France.
| | | |
Collapse
|
159
|
Tsuboi K, Ueda N. [Enzymes involved in the degradation of N-acylethanolamines functioning as lipid mediators]. Nihon Yakurigaku Zasshi 2011; 138:8-12. [PMID: 21747202 DOI: 10.1254/fpj.138.8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
|
160
|
Scott CW, Tian G, Yu XH, Paschetto KA, Wilkins DE, Meury L, Cao CQ, Varnes J, Edwards PD. Biochemical characterization and in vitro activity of AZ513, a noncovalent, reversible, and noncompetitive inhibitor of fatty acid amide hydrolase. Eur J Pharmacol 2011; 667:74-9. [PMID: 21645511 DOI: 10.1016/j.ejphar.2011.05.052] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2011] [Revised: 05/16/2011] [Accepted: 05/23/2011] [Indexed: 10/18/2022]
Abstract
Fatty acid amide hydrolase (FAAH) hydrolyzes several bioactive lipids including the endocannabinoid anandamide. Synthetic FAAH inhibitors are being generated to help define the biological role(s) of this enzyme, the lipids it degrades in vivo, and the disease states that might benefit from its pharmacological modulation. AZ513 inhibits human FAAH (IC(50)=551 nM), is 20-fold more potent against rat FAAH (IC(50)=27 nM), and is inactive at 10 μM against the serine hydrolases acetylcholinesterase, thrombin, and trypsin. In contrast to most other potent FAAH inhibitors, AZ513 showed no evidence of covalently modifying the enzyme and displayed reversible inhibition. In an enzyme cross-competition assay, AZ513 did not compete with OL-135, an inhibitor that binds to the catalytic site in FAAH, which indicates that AZ513 does not bind to the catalytic site and is therefore noncompetitive with respect to substrate. AZ513 has good cell penetration as demonstrated by inhibition of anandamide hydrolysis in human FAAH-transfected HEK293 cells (IC(50)=360 nM). AZ513 was tested in a rat spinal cord slice preparation where CB(1) activation reduces excitatory post-synaptic currents (EPSCs). In this native tissue assay of synaptic activity, AZ513 reduced EPSCs, which is consistent with inhibiting endogenous FAAH and augmenting endocannabinoid tone. AZ513 has a unique biochemical profile compared with other published FAAH inhibitors and will be a useful tool compound to further explore the role of FAAH in various biological processes.
Collapse
Affiliation(s)
- Clay W Scott
- CNS Discovery Research, AstraZeneca Pharmaceuticals, 1800 Concord Pike, Wilmington, DE 19803, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
161
|
Wilson AA, Garcia A, Parkes J, Houle S, Tong J, Vasdev N. [11C]CURB: Evaluation of a novel radiotracer for imaging fatty acid amide hydrolase by positron emission tomography. Nucl Med Biol 2011; 38:247-53. [PMID: 21315280 DOI: 10.1016/j.nucmedbio.2010.08.001] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2010] [Revised: 08/05/2010] [Accepted: 08/08/2010] [Indexed: 01/02/2023]
Abstract
INTRODUCTION Fatty acid amide hydrolase (FAAH) is the enzyme responsible for metabolising the endogenous cannabinoid, anandamide, and thus represents an important target for molecular imaging. To date, no radiotracer has been shown to be useful for imaging of FAAH using either positron emission tomography (PET) or single photon emission computed tomography (SPECT). We here determine the suitability of a novel carbon-11-labeled inhibitor of FAAH via ex vivo biodistribution studies in rat brain in conjunction with pharmacological challenges. METHODS A potent irreversible inhibitor of FAAH, URB694, radiolabeled with carbon-11 in the carbonyl position ([(11)C]CURB), was administered to male rats via tail-vein injection. Rats were sacrificed at various time points postinjection, and tissue samples were dissected, counted and weighed. Specific binding to FAAH was investigated by pretreatment of animals with URB694 or URB597. For metabolism and mechanism of binding studies, whole brains were excised post-radiotracer injection, homogenised and extracted exhaustively with 80% aq. acetonitrile to determine the time course and fraction of radioactivity that was irreversibly bound to brain parenchyma. RESULTS Upon intravenous injection into rats, [(11)C]CURB showed high brain uptake [standard uptake value (SUV) of 1.6-2.4 at 5 min] with little washout over time, which is characteristic of irreversible binding. Highest uptake of radioactivity was seen in the cortex, intermediate in the cerebellum and lowest in the hypothalamus, reflecting the reported distribution of FAAH. Brain uptake of radioactivity was decreased in a dose-dependent manner by pretreatment with increasing amounts of URB694, demonstrating that binding was saturable. Pretreatment with the well-characterised FAAH inhibitor, URB597, reduced binding in all brain regions by 70-80%. Homogenised brain extraction experiments demonstrated unequivocally that [(11)C]CURB was irreversibly bound to FAAH. CONCLUSIONS The title radiotracer demonstrates favourable properties such as good brain uptake, regional heterogeneity and specificity of binding based on ex vivo biodistribution studies in conscious rat brain. [(11)C]CURB represents a highly promising radiotracer for the imaging of FAAH using PET.
Collapse
Affiliation(s)
- Alan A Wilson
- PET Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada.
| | | | | | | | | | | |
Collapse
|
162
|
Ezzili C, Mileni M, McGlinchey N, Long JZ, Kinsey SG, Hochstatter DG, Stevens RC, Lichtman AH, Cravatt BF, Bilsky EJ, Boger DL. Reversible competitive α-ketoheterocycle inhibitors of fatty acid amide hydrolase containing additional conformational constraints in the acyl side chain: orally active, long-acting analgesics. J Med Chem 2011; 54:2805-22. [PMID: 21428410 PMCID: PMC3085948 DOI: 10.1021/jm101597x] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
A series of α-ketooxazoles containing conformational constraints in the C2 acyl side chain of 2 (OL-135) were examined as inhibitors of fatty acid amide hydrolase (FAAH). Only one of the two possible enantiomers displayed potent FAAH inhibition (S vs R enantiomer), and their potency is comparable or improved relative to 2, indicating that the conformational restriction in the C2 acyl side chain is achievable. A cocrystal X-ray structure of the α-ketoheterocycle 12 bound to a humanized variant of rat FAAH revealed its binding details, confirmed that the (S)-enantiomer is the bound active inhibitor, shed light on the origin of the enantiomeric selectivity, and confirmed that the catalytic Ser241 is covalently bound to the electrophilic carbonyl as a deprotonated hemiketal. Preliminary in vivo characterization of the inhibitors 12 and 14 is reported demonstrating that they raise brain anandamide levels following either intraperitoneal (ip) or oral (po) administration indicative of effective in vivo FAAH inhibition. Significantly, the oral administration of 12 caused dramatic accumulation of anandamide in the brain, with peak levels achieved between 1.5 and 3 h, and these elevations were maintained over 9 h. Additional studies of these two representative members of the series (12 and 14) in models of thermal hyperalgesia and neuropathic pain are reported, including the demonstration that 12 administered orally significantly attenuated mechanical (>6 h) and cold (>9 h) allodynia for sustained periods consistent with its long-acting effects in raising the endogenous concentration of anandamide.
Collapse
Affiliation(s)
- Cyrine Ezzili
- Department of Chemistry, 10550 North Torrey Pines Road, La Jolla, California 92037
| | - Mauro Mileni
- Department of Molecular Biology, 10550 North Torrey Pines Road, La Jolla, California 92037
| | - Nicholas McGlinchey
- Department of Pharmacology, College of Osteopathic Medicine, University of New England, Biddeford, ME 04005
| | - Jonathan Z. Long
- Department of Chemical Physiology, 10550 North Torrey Pines Road, La Jolla, California 92037
| | - Steven G. Kinsey
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA 23298
| | | | - Raymond C. Stevens
- Department of Molecular Biology, 10550 North Torrey Pines Road, La Jolla, California 92037
- The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037
| | - Aron H. Lichtman
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA 23298
| | - Benjamin F. Cravatt
- Department of Chemical Physiology, 10550 North Torrey Pines Road, La Jolla, California 92037
- The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037
| | - Edward J. Bilsky
- Department of Pharmacology, College of Osteopathic Medicine, University of New England, Biddeford, ME 04005
| | - Dale L. Boger
- Department of Chemistry, 10550 North Torrey Pines Road, La Jolla, California 92037
- The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037
| |
Collapse
|
163
|
Discovery and molecular basis of potent noncovalent inhibitors of fatty acid amide hydrolase (FAAH). Proc Natl Acad Sci U S A 2011; 108:7379-84. [PMID: 21502526 DOI: 10.1073/pnas.1016167108] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Fatty acid amide hydrolase (FAAH), an amidase-signature family member, is an integral membrane enzyme that degrades lipid amides including the endogenous cannabinoid anandamide and the sleep-inducing molecule oleamide. Both genetic knock out and pharmacological administration of FAAH inhibitors in rodent models result in analgesic, anxiolytic, and antiinflammatory phenotypes. Targeting FAAH activity, therefore, presents a promising new therapeutic strategy for the treatment of pain and other neurological-related or inflammatory disorders. Nearly all FAAH inhibitors known to date attain their binding potency through a reversible or irreversible covalent modification of the nucleophile Ser241 in the unusual Ser-Ser-Lys catalytic triad. Here, we report the discovery and mechanism of action of a series of ketobenzimidazoles as unique and potent noncovalent FAAH inhibitors. Compound 2, a representative of these ketobenzimidazoles, was designed from a series of ureas that were identified from high-throughput screening. While urea compound 1 is characterized as an irreversible covalent inhibitor, the cocrystal structure of FAAH complexed with compound 2 reveals that these ketobenzimidazoles, though containing a carbonyl moiety, do not covalently modify Ser241. These inhibitors achieve potent inhibition of FAAH activity primarily from shape complementarity to the active site and through numerous hydrophobic interactions. These noncovalent compounds exhibit excellent selectivity and good pharmacokinetic properties. The discovery of this distinctive class of inhibitors opens a new avenue for modulating FAAH activity through nonmechanism-based inhibition.
Collapse
|
164
|
Mileni M, Garfunkle J, Ezzili C, Cravatt BF, Stevens RC, Boger DL. Fluoride-mediated capture of a noncovalent bound state of a reversible covalent enzyme inhibitor: X-ray crystallographic analysis of an exceptionally potent α-ketoheterocycle inhibitor of fatty acid amide hydrolase. J Am Chem Soc 2011; 133:4092-100. [PMID: 21355555 PMCID: PMC3060301 DOI: 10.1021/ja110877y] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Two cocrystal X-ray structures of the exceptionally potent α-ketoheterocycle inhibitor 1 (K(i) = 290 pM) bound to a humanized variant of rat fatty acid amide hydrolase (FAAH) are disclosed, representing noncovalently and covalently bound states of the same inhibitor with the enzyme. Key to securing the structure of the noncovalently bound state of the inhibitor was the inclusion of fluoride ion in the crystallization conditions that is proposed to bind the oxyanion hole precluding inhibitor covalent adduct formation with stabilization of the tetrahedral hemiketal. This permitted the opportunity to detect important noncovalent interactions stabilizing the binding of the inhibitor within the FAAH active site independent of the covalent reaction. Remarkably, noncovalently bound 1 in the presence of fluoride appears to capture the active site in the same "in action" state with the three catalytic residues Ser241-Ser217-Lys142 occupying essentially identical positions observed in the covalently bound structure of 1, suggesting that this technique of introducing fluoride may have important applications in structural studies beyond inhibiting substrate or inhibitor oxyanion hole binding. Key insights to emerge from the studies include the observations that noncovalently bound 1 binds in its ketone (not gem diol) form, that the terminal phenyl group in the acyl side chain of the inhibitor serves as the key anchoring interaction overriding the intricate polar interactions in the cytosolic port, and that the role of the central activating heterocycle is dominated by its intrinsic electron-withdrawing properties. These two structures are also briefly compared with five X-ray structures of α-ketoheterocycle-based inhibitors bound to FAAH recently disclosed.
Collapse
Affiliation(s)
- Mauro Mileni
- Department of Molecular Biology, The Scripps Research
Institute, 10550 North Torrey Pines Road, La Jolla, California 92037
| | - Joie Garfunkle
- Department of Chemistry, The Scripps Research Institute,
10550 North Torrey Pines Road, La Jolla, California 92037
- The Skaggs Institute for Chemical Biology, The Scripps
Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037
| | - Cyrine Ezzili
- Department of Chemistry, The Scripps Research Institute,
10550 North Torrey Pines Road, La Jolla, California 92037
- The Skaggs Institute for Chemical Biology, The Scripps
Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037
| | - Benjamin F. Cravatt
- Department of Chemical Physiology, The Scripps Research
Institute, 10550 North Torrey Pines Road, La Jolla, California 92037
- The Skaggs Institute for Chemical Biology, The Scripps
Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037
| | - Raymond C. Stevens
- Department of Chemistry, The Scripps Research Institute,
10550 North Torrey Pines Road, La Jolla, California 92037
- Department of Molecular Biology, The Scripps Research
Institute, 10550 North Torrey Pines Road, La Jolla, California 92037
| | - Dale L. Boger
- Department of Chemistry, The Scripps Research Institute,
10550 North Torrey Pines Road, La Jolla, California 92037
- The Skaggs Institute for Chemical Biology, The Scripps
Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037
| |
Collapse
|
165
|
Capoferri L, Mor M, Sirirak J, Chudyk E, Mulholland AJ, Lodola A. Application of a SCC-DFTB QM/MM approach to the investigation of the catalytic mechanism of fatty acid amide hydrolase. J Mol Model 2011; 17:2375-83. [PMID: 21365225 DOI: 10.1007/s00894-011-0981-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2010] [Accepted: 01/21/2011] [Indexed: 12/20/2022]
Abstract
Self-consistent charge density functional tight binding (SCC-DFTB) is a promising method for hybrid quantum mechanics/molecular mechanics (QM/MM) simulations of enzyme-catalyzed reactions. The acylation reaction of fatty acid amide hydrolase (FAAH), a promising drug target, was investigated by applying a SCC-DFTB/CHARMM27 scheme. Calculated potential energy barriers resulted in reasonable agreement with experiments for oleamide (OA) and oleoylmethyl ester (OME) substrates, outperforming previous calculations performed at the PM3/CHARMM22 level. Furthermore, the experimental preference of FAAH in hydrolyzing OA faster than OME was adequately reproduced by calculations. All these findings indicate that the SCC-DFTB/CHARMM27 approach can be successfully applied to mechanistic investigations of FAAH-catalyzed reactions.
Collapse
Affiliation(s)
- Luigi Capoferri
- Dipartimento Farmaceutico, Università degli Studi di Parma, viale G. P. Usberti 27/A Campus Universitario, 43124, Parma, Italy
| | | | | | | | | | | |
Collapse
|
166
|
Application of Computational Methods to the Design of Fatty Acid Amide Hydrolase (FAAH) Inhibitors Based on a Carbamic Template Structure. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2011; 85:1-26. [DOI: 10.1016/b978-0-12-386485-7.00001-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/05/2023]
|
167
|
Wang Y, Jones P. A scintillation proximity assay for fatty acid amide hydrolase compatible with inhibitor screening. Methods Mol Biol 2010; 572:247-59. [PMID: 20694697 DOI: 10.1007/978-1-60761-244-5_16] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/19/2023]
Abstract
Scintillation proximity assay (SPA) is a homogenous and versatile technology for the simple and sensitive detection of the interaction of protein targets with their ligands. Herein, we described a SPA assay developed to identify compounds that bind to human fatty acid amide hydrolase (FAAH). This SPA assay utilizes the specific binding of [(3)H]-R(+)-methanandamide ((3)H-MAEA), a competitive nonhydrolyzed FAAH inhibitor, to FAAH expressing microsomes and evaluates its displacement by FAAH inhibitors. In contrast to the classical SPA radioligand binding assay which detects bound ligand, in our assay the released radiolabel is detected through its interaction with the SPA beads. This novel SPA assay has been validated and demonstrated to be simple, sensitive, and amenable to high-throughput screening.
Collapse
Affiliation(s)
- Yuren Wang
- Neuroscience Discovery Research, Wyeth Research, Princeton, NJ, USA
| | | |
Collapse
|
168
|
Albury MS, Elliott C, Moore AL. Ubiquinol-binding site in the alternative oxidase: Mutagenesis reveals features important for substrate binding and inhibition. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2010; 1797:1933-9. [DOI: 10.1016/j.bbabio.2010.01.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2009] [Revised: 01/11/2010] [Accepted: 01/12/2010] [Indexed: 11/16/2022]
|
169
|
The electron transfer flavoprotein: ubiquinone oxidoreductases. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2010; 1797:1910-6. [PMID: 20937244 DOI: 10.1016/j.bbabio.2010.10.007] [Citation(s) in RCA: 170] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2010] [Revised: 07/20/2010] [Accepted: 10/05/2010] [Indexed: 12/30/2022]
Abstract
Electron transfer flavoprotein: ubiqionone oxidoreductase (ETF-QO) is a component of the mitochondrial respiratory chain that together with electron transfer flavoprotein (ETF) forms a short pathway that transfers electrons from 11 different mitochondrial flavoprotein dehydrogenases to the ubiquinone pool. The X-ray structure of the pig liver enzyme has been solved in the presence and absence of a bound ubiquinone. This structure reveals ETF-QO to be a monotopic membrane protein with the cofactors, FAD and a [4Fe-4S](+1+2) cluster, organised to suggests that it is the flavin that serves as the immediate reductant of ubiquinone. ETF-QO is very highly conserved in evolution and the recombinant enzyme from the bacterium Rhodobacter sphaeroides has allowed the mutational analysis of a number of residues that the structure suggested are involved in modulating the reduction potential of the cofactors. These experiments, together with the spectroscopic measurement of the distances between the cofactors in solution have confirmed the intramolecular pathway of electron transfer from ETF to ubiquinone. This approach can be extended as the R. sphaeroides ETF-QO provides a template for investigating the mechanistic consequences of single amino acid substitutions of conserved residues that are associated with a mild and late onset variant of the metabolic disease multiple acyl-CoA dehydrogenase deficiency (MADD).
Collapse
|
170
|
Abstract
N-acylethanolamides (NAEs) are naturally occurring signaling lipids consisting of amides and esters of long-chain polyunsaturated fatty acids. Usually they are present in a very small amounts in many mammalian tissues and cells, including human reproductive tracts and fluids. Recently, the presence of N-arachidonoylethanolamide (anandamide, AEA), the most characterised member of endocannabinoids, and its congeners palmitoylethanolamide (PEA) and oleylethanolamide (OEA) in seminal plasma, oviductal fluid, and follicular fluids was demonstrated. AEA has been shown to bind not only type-1 (CB1) and type-2 (CB2) cannabinoid receptors, but also type-1 vanilloid receptor (TRPV1), while PEA and OEA are inactive with respect to classical cannabinoid CB1 and CB2 but activate TRPV1 or peroxisome proliferator activate receptors (PPARs). This review concerns the most recent experimental data on PEA and OEA, endocannabinoid-like molecules which appear to exert their action exclusively on sperm cells with altered features, such as membrane characteristics and kinematic parameters. Their beneficial effects on these cells could suggest a possible pharmacological use of PEA and OEA on patients affected by some forms of idiopathic infertility.
Collapse
|
171
|
Hu SSJ, Arnold A, Hutchens JM, Radicke J, Cravatt BF, Wager-Miller J, Mackie K, Straiker A. Architecture of cannabinoid signaling in mouse retina. J Comp Neurol 2010; 518:3848-66. [PMID: 20653038 PMCID: PMC2982216 DOI: 10.1002/cne.22429] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Cannabinoid receptors and their ligands constitute an endogenous signaling system that is found throughout the body, including the eye. This system can be activated by Delta(9)-tetrahydrocannabinol, a major drug of abuse. Cannabinoids offer considerable therapeutic potential in modulating ocular immune and inflammatory responses and in regulating intraocular pressure. The location of cannabinoid receptor 1 (CB(1)) in the retina is known, but recently a constellation of proteins has been identified that produce and break down endocannabinoids (eCBs) and modulate CB(1) function. Localization of these proteins is critical to defining specific cannabinoid signaling circuitry in the retina. Here we show the localization of diacylglycerol lipase-alpha and -beta (DGLalpha/beta), implicated in the production of the eCB 2-arachidonoyl glycerol (2-AG); monoacylglycerol lipase (MGL) and alpha/beta-hydrolase domain 6 (ABHD6), both implicated in the breakdown of 2-AG; cannabinoid receptor-interacting protein 1a (CRIP1a), a protein that may modulate CB(1) function; and fatty acid amide hydrolase (FAAH) and N-acylethanolamine-hydrolyzing acid amidase (NAAA), which have been shown to break down the eCB anandamide and related acyl amides. Our most prominent finding was that DGLalpha is present in postsynaptic type 1 OFF cone bipolar cells juxtaposed to CB(1)-containing cone photoreceptor terminals. CRIP1a is reliably presynaptic to DGLalpha, consistent with a possible role in cannabinoid signaling, and NAAA is restricted to retinal pigment epithelium, whereas DGLbeta is limited to retinal blood vessels. These results taken together with previous anatomical and functional studies define specific cannabinoid circuitry likely to modulate eCB signaling at the first synapse of the retina as well as in the inner plexiform layer.
Collapse
Affiliation(s)
- Sherry Shu-Jung Hu
- The Gill Center for Biomolecular Science and the Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN 47405, USA
| | - Andy Arnold
- The Gill Center for Biomolecular Science and the Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN 47405, USA
| | - Jacqueline M. Hutchens
- The Gill Center for Biomolecular Science and the Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN 47405, USA
| | - Josh Radicke
- The Gill Center for Biomolecular Science and the Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN 47405, USA
| | - Benjamin F. Cravatt
- Department of Chemical Physiology, the Scripps Research Institute, La Jolla, CA 92037, USA
| | - Jim Wager-Miller
- The Gill Center for Biomolecular Science and the Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN 47405, USA
| | - Ken Mackie
- The Gill Center for Biomolecular Science and the Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN 47405, USA
| | - Alex Straiker
- The Gill Center for Biomolecular Science and the Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN 47405, USA
| |
Collapse
|
172
|
Deng H. Recent advances in the discovery and evaluation of fatty acid amide hydrolase inhibitors. Expert Opin Drug Discov 2010; 5:961-93. [PMID: 22823990 DOI: 10.1517/17460441.2010.513378] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
IMPORTANCE OF THE FIELD Cannabis has been used for both medicinal and recreational purposes since ancient times. Although cannabinoid-based medicines hold great promise in several challenging therapeutic areas such as pain management and mode control, their development has been hampered by psychoactive and other CNS-related side effects. The identification of fatty acid amide hydrolase (FAAH), a key enzyme responsible for the degradation of endocannabinoids, has brought in tremendous opportunities in that inhibition of FAAH leads to local elevation of endocannabinoids under certain stimuli, thus, avoiding the side effects from global activation of cannabinoid receptors by exogenous cannabimimetic compounds. The search for selective FAAH inhibitors has thus become a strong focus in current drug discovery. AREAS COVERED IN THIS REVIEW This review summarizes our current understanding of FAAH including its structure, catalytic mechanism and biological functions with emphases on its role in the regulation of endocannabinoids and other signaling lipids. The review then highlights the most recent discovery and biological activities of different classes of FAAH inhibitors. Last, the review discusses challenges and potential drawbacks in the development of FAAH inhibitor-based therapy. WHAT THE READER WILL GAIN Readers will have an overview of FAAH and obtain a rationale on FAAH as an attractive therapeutic target for the development of medicines for treating pain, inflammation, anxiety and other diseases. More importantly, readers will gain knowledge on various newly established FAAH inhibitor scaffolds and their development potentials, and such information will hopefully stimulate ideas for the designing of new inhibitors with superior activity profiles. The discussions on the potential challenges in developing FAAH inhibitors will impose more caution in the decision-making process, thus, lowering the possibility of late stage failure. TAKE HOME MESSAGE FAAH is an attractive target for modulating the endocannabinoid system, thus, treating many disease conditions including pain and mode control without the CNS side effects associated with cannabis usage. In recent years, tremendous effort has been focused in the FAAH inhibitor research field, and consequently many novel chemical templates have been discovered. FAAH hydrolyzes several important signaling lipids, but the long-term effects of FAAH inhibition in humans remain to be seen. While it is challenging to identify the right molecule with the right level of intervention of the FAAH function for treating a disease condition, it is possible to avoid mechanism-related undesired effects. With the entry of several compounds into clinical trials, FAAH inhibitor-based medicines are on the horizon.
Collapse
Affiliation(s)
- Hongfeng Deng
- GlaxoSmithKline, Platform Technology & Science/Molecular Discovery Research, ELT-Boston, 830 Winter Street, Waltham, MA 02451, USA
| |
Collapse
|
173
|
Ueda N, Tsuboi K, Uyama T. Enzymological studies on the biosynthesis of N-acylethanolamines. Biochim Biophys Acta Mol Cell Biol Lipids 2010; 1801:1274-85. [PMID: 20736084 DOI: 10.1016/j.bbalip.2010.08.010] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2010] [Revised: 08/10/2010] [Accepted: 08/17/2010] [Indexed: 11/28/2022]
Abstract
Ethanolamides of different long-chain fatty acids constitute a class of endogenous lipid molecules generally called N-acylethanolamines (NAEs). They contain N-arachidonoylethanolamine (anandamide), N-palmitoylethanolamine, and N-oleoylethanolamine, which receive considerable attention because of their actions as an endogenous cannabinoid receptor ligand (endocannabinoid), an anti-inflammatory substance, and an appetite-suppressing substance, respectively. Identification of their biosynthetic routes in animal tissues and molecular characterization of the enzymes involved are essential for better understanding of physiological importance of NAEs as well as development of enzyme inhibitors as possible therapeutic drugs. In the classical "transacylation-phosphodiesterase pathway", NAEs are formed from glycerophospholipids via N-acylphosphatidylethanolamine (NAPE), an unusual derivative of phosphatidylethanolamine with a third acyl chain attached to the amino group, by sequential catalyses by Ca(2+)-dependent N-acyltransferase and NAPE-hydrolyzing phospholipase D. However, recent studies reveal that NAE-generating pathways are more complex than presumed before. In this review article, we will focus on recent findings regarding mammalian enzymes that are involved or might be involved in the biosynthesis of NAEs.
Collapse
Affiliation(s)
- Natsuo Ueda
- Department of Biochemistry, Kagawa University School of Medicine, 1750-1 Ikenobe, Miki, Kagawa 761-0793, Japan.
| | | | | |
Collapse
|
174
|
Indole-3-acetamide-dependent auxin biosynthesis: a widely distributed way of indole-3-acetic acid production? Eur J Cell Biol 2010; 89:895-905. [PMID: 20701997 DOI: 10.1016/j.ejcb.2010.06.021] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
During the course of evolution plants have evolved a complex phytohormone-based network to regulate their growth and development. Herein auxins have a pivotal function, as they are involved in controlling virtually every aspect related to plant growth. Indole-3-acetic acid (IAA) is the major endogenous auxin of higher plants that is already known for more than 80 years. In spite of the long-standing interest in this topic, IAA biosynthesis is still only partially uncovered. Several pathways for the formation of IAA have been proposed over the past years, but none of these pathways are yet completely defined. The aim of this review is to summarize the current knowledge on the indole-3-acetamide (IAM)-dependent pathway of IAA production in plants and to discuss the properties of the involved proteins and genes, respectively. Their evolutionary relationship to known bacterial IAM hydrolases and other amidases from bacteria, algae, moss, and higher plants is discussed on the basis of phylogenetic analyses. Moreover, we report on the transcriptional regulation of the Arabidopsis AMI1 gene.
Collapse
|
175
|
Lodola A, Sirirak J, Fey N, Rivara S, Mor M, Mulholland AJ. Structural Fluctuations in Enzyme-Catalyzed Reactions: Determinants of Reactivity in Fatty Acid Amide Hydrolase from Multivariate Statistical Analysis of Quantum Mechanics/Molecular Mechanics Paths. J Chem Theory Comput 2010; 6:2948-60. [PMID: 26616091 DOI: 10.1021/ct100264j] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The effects of structural fluctuations, due to protein dynamics, on enzyme activity are at the heart of current debates on enzyme catalysis. There is evidence that fatty acid amide hydrolase (FAAH) is an enzyme for which reaction proceeds via a high-energy, reactive conformation, distinct from the predominant enzyme-substrate complex (Lodola et al. Biophys. J. 2007, 92, L20-22). Identifying the structural causes of differences in reactivity between conformations in such complex systems is not trivial. Here, we show that multivariate analysis of key structural parameters can identify structural determinants of barrier height by analysis of multiple reaction paths. We apply a well-tested quantum mechanics/molecular mechanics (QM/MM) method to the first step of the acylation reaction between FAAH and oleamide substrate for 36 different starting structures. Geometrical parameters (consisting of the key bond distances that change during the reaction) were collected and used for principal component analysis (PCA), partial least-squares (PLS) regression analysis, and multiple linear regression (MLR) analysis. PCA indicates that different "families" of enzyme-substrate conformations arise from QM/MM molecular dynamics simulation and that rarely sampled, catalytically significant conformational states can be identified. PLS and MLR analyses allowed the construction of linear regression models, correlating the calculated activation barriers with simple geometrical descriptors. These analyses reveal the presence of two fully independent geometrical effects, explaining 78% of the variation in the activation barrier, which are directly correlated with transition-state stabilization (playing a major role in catalysis) and substrate binding. These results highlight the power of statistical approaches of this type in identifying crucial structural features that contribute to enzyme reactivity.
Collapse
Affiliation(s)
- Alessio Lodola
- Dipartimento Farmaceutico, Università degli Studi di Parma, 43124 Parma, Italy, and Centre for Computational Chemistry, School of Chemistry, University of Bristol, Bristol BS8 1TS, United Kingdom
| | - Jitnapa Sirirak
- Dipartimento Farmaceutico, Università degli Studi di Parma, 43124 Parma, Italy, and Centre for Computational Chemistry, School of Chemistry, University of Bristol, Bristol BS8 1TS, United Kingdom
| | - Natalie Fey
- Dipartimento Farmaceutico, Università degli Studi di Parma, 43124 Parma, Italy, and Centre for Computational Chemistry, School of Chemistry, University of Bristol, Bristol BS8 1TS, United Kingdom
| | - Silvia Rivara
- Dipartimento Farmaceutico, Università degli Studi di Parma, 43124 Parma, Italy, and Centre for Computational Chemistry, School of Chemistry, University of Bristol, Bristol BS8 1TS, United Kingdom
| | - Marco Mor
- Dipartimento Farmaceutico, Università degli Studi di Parma, 43124 Parma, Italy, and Centre for Computational Chemistry, School of Chemistry, University of Bristol, Bristol BS8 1TS, United Kingdom
| | - Adrian J Mulholland
- Dipartimento Farmaceutico, Università degli Studi di Parma, 43124 Parma, Italy, and Centre for Computational Chemistry, School of Chemistry, University of Bristol, Bristol BS8 1TS, United Kingdom
| |
Collapse
|
176
|
Mileni M, Kamtekar S, Wood DC, Benson TE, Cravatt BF, Stevens RC. Crystal structure of fatty acid amide hydrolase bound to the carbamate inhibitor URB597: discovery of a deacylating water molecule and insight into enzyme inactivation. J Mol Biol 2010; 400:743-54. [PMID: 20493882 PMCID: PMC3014312 DOI: 10.1016/j.jmb.2010.05.034] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2010] [Revised: 05/13/2010] [Accepted: 05/14/2010] [Indexed: 12/22/2022]
Abstract
The endocannabinoid system regulates a wide range of physiological processes including pain, inflammation, and cognitive/emotional states. URB597 is one of the best characterized covalent inhibitors of the endocannabinoid-degrading enzyme fatty acid amide hydrolase (FAAH). Here, we report the structure of the FAAH-URB597 complex at 2.3 A resolution. The structure provides insights into mechanistic details of enzyme inactivation and experimental evidence of a previously uncharacterized active site water molecule that likely is involved in substrate deacylation. This water molecule is part of an extensive hydrogen-bonding network and is coordinated indirectly to residues lining the cytosolic port of the enzyme. In order to corroborate our hypothesis concerning the role of this water molecule in FAAH's catalytic mechanism, we determined the structure of FAAH conjugated to a urea-based inhibitor, PF-3845, to a higher resolution (2.4 A) than previously reported. The higher-resolution structure confirms the presence of the water molecule in a virtually identical location in the active site. Examination of the structures of serine hydrolases that are non-homologous to FAAH, such as elastase, trypsin, or chymotrypsin, shows a similarly positioned hydrolytic water molecule and suggests a functional convergence between the amidase signature enzymes and serine proteases.
Collapse
Affiliation(s)
- Mauro Mileni
- Department of Molecular Biology, The Scripps Research Institute, La Jolla, CA 92037 USA
| | - Satwik Kamtekar
- Pfizer Global Research and Development, Chesterfield, MO 63017, USA
| | - David C. Wood
- Pfizer Global Research and Development, Chesterfield, MO 63017, USA
| | | | - Benjamin F. Cravatt
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA 92037 USA
| | - Raymond C. Stevens
- Department of Molecular Biology, The Scripps Research Institute, La Jolla, CA 92037 USA
| |
Collapse
|
177
|
Papahatjis DP, Nahmias VR, Nikas SP, Schimpgen M, Makriyannis A. Design and synthesis of (13S)-methyl-substituted arachidonic acid analogues: templates for novel endocannabinoids. Chemistry 2010; 16:4091-9. [PMID: 20187040 DOI: 10.1002/chem.200902880] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Two novel methyl-substituted arachidonic acid derivatives were prepared in an enantioselective manner from commercially available chiral building blocks, and were found to be excellent templates for the development of (13S)-methyl-substituted anandamide analogues. One of the compounds synthesized, namely, (13S,5Z,8Z,11Z,14Z)-13-methyl-eicosa-5,8,11,14-tetraenoic acid N-(2-hydroxyethyl)amide, is an endocannabinoid analogue with remarkably high affinity for the CB1 cannabinoid receptor.
Collapse
Affiliation(s)
- Demetris P Papahatjis
- Institute of Organic and Pharmaceutical Chemistry, National Hellenic Research Foundation, 48 Vass. Constantinou, Athens 116-35, Greece.
| | | | | | | | | |
Collapse
|
178
|
A new group of oxime carbamates as reversible inhibitors of fatty acid amide hydrolase. Bioorg Med Chem Lett 2010; 20:4406-11. [PMID: 20591666 DOI: 10.1016/j.bmcl.2010.06.050] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2010] [Revised: 06/09/2010] [Accepted: 06/09/2010] [Indexed: 11/23/2022]
Abstract
A series of oxime carbamates have been identified as potent inhibitors of fatty acid amide hydrolase (FAAH), an important regulatory enzyme of the endocannabinoid signaling system. Kinetic analysis indicates that they behave as non-competitive, reversible inhibitors, and show remarkable selectivity for FAAH over the other components of the endocannabinoid system.
Collapse
|
179
|
Li S, Zhang X, Wang W. Cluster formation of anchored proteins induced by membrane-mediated interaction. Biophys J 2010; 98:2554-63. [PMID: 20513399 PMCID: PMC2877327 DOI: 10.1016/j.bpj.2010.02.032] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2009] [Revised: 01/16/2010] [Accepted: 02/17/2010] [Indexed: 11/16/2022] Open
Abstract
Computer simulations were used to study the cluster formation of anchored proteins in a membrane. The rate and extent of clustering was found to be dependent upon the hydrophobic length of the anchored proteins embedded in the membrane. The cluster formation mechanism of anchored proteins in our work was ascribed to the different local perturbations on the upper and lower monolayers of the membrane and the intermonolayer coupling. Simulation results demonstrated that only when the penetration depth of anchored proteins was larger than half the membrane thickness, could the structure of the lower monolayer be significantly deformed. Additionally, studies on the local structures of membranes indicated weak perturbation of bilayer thickness for a shallowly inserted protein, while there was significant perturbation for a more deeply inserted protein. The origin of membrane-mediated protein-protein interaction is therefore due to the local perturbation of the membrane thickness, and the entropy loss-both of which are caused by the conformation restriction on the lipid chains and the enhanced intermonolayer coupling for a deeply inserted protein. Finally, in this study we addressed the difference of cluster formation mechanisms between anchored proteins and transmembrane proteins.
Collapse
Affiliation(s)
| | - Xianren Zhang
- Division of Molecular and Materials Simulation, Key Laboratory for Nanomaterials, Ministry of Education, Beijing University of Chemical Technology, Beijing, China
| | | |
Collapse
|
180
|
Petrosino S, Cristino L, Karsak M, Gaffal E, Ueda N, Tüting T, Bisogno T, De Filippis D, D'Amico A, Saturnino C, Orlando P, Zimmer A, Iuvone T, Di Marzo V. Protective role of palmitoylethanolamide in contact allergic dermatitis. Allergy 2010; 65:698-711. [PMID: 19909294 DOI: 10.1111/j.1398-9995.2009.02254.x] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
BACKGROUND Palmitoylethanolamide (PEA) is an anti-inflammatory mediator that enhances the activation by anandamide (AEA) of cannabinoid receptors and transient receptor potential vanilloid type-1 (TRPV1) channels, and directly activates peroxisome proliferator-activated receptor-alpha (PPAR-alpha). In mice, 2,4-dinitrofluorobenzene (DNFB)-induced contact allergic dermatitis (CAD) in inflamed ears is partly mediated by the chemokine Monocyte Chemotactic Protein-2 (MCP-2) and accompanied by elevation of AEA levels. No datum is available on PEA regulation and role in CAD. OBJECTIVE We examined whether PEA is produced during DNFB-induced CAD, and if it has any direct protective action in keratinocytes in vitro. METHODS Eight- to ten-week-old female C57BL/6J wild-type and CB(1)/CB(2) double knock-out mice were used to measure PEA levels and the expression of TRPV1, PPAR-alpha receptors and enzymes responsible for PEA biosynthesis and degradation. Human keratinocytes (HaCaT) cells were stimulated with polyinosinic polycytidylic acid [poly-(I:C)], and the expression and release of MCP-2 were measured in the presence of PEA and antagonists of its proposed receptors. RESULTS 2,4-Dinitrofluorobenzene increased ear skin PEA levels and up-regulated TRPV1, PPAR-alpha and a PEA-biosynthesizing enzyme in ear keratinocytes. In HaCaT cells, stimulation with poly-(I:C) elevated the levels of both PEA and AEA, and exogenous PEA (10 microM) inhibited poly-(I:C)-induced expression and release of MCP-2 in a way reversed by antagonism at TRPV1, but not PPAR-alpha. PEA (5-10 mg/kg, intraperitoneal) also inhibited DNFB-induced ear inflammation in mice in vivo, in a way attenuated by TRPV1 antagonism. CONCLUSIONS We suggest that PEA is an endogenous protective agent against DNFB-induced keratinocyte inflammation and could be considered for therapeutic use against CAD.
Collapse
Affiliation(s)
- S Petrosino
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry, Consiglio Nazionale delle Ricerche, Pozzuoli (Naples), Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
181
|
Freigang S, Zadorozhny V, McKinney MK, Krebs P, Herro R, Pawlak J, Kain L, Schrantz N, Masuda K, Liu Y, Savage PB, Bendelac A, Cravatt BF, Teyton L. Fatty acid amide hydrolase shapes NKT cell responses by influencing the serum transport of lipid antigen in mice. J Clin Invest 2010; 120:1873-84. [PMID: 20484813 PMCID: PMC2877940 DOI: 10.1172/jci40451] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2009] [Accepted: 03/31/2010] [Indexed: 11/17/2022] Open
Abstract
The potent regulatory properties of NKT cells render this subset of lipid-specific T cells a promising target for immunotherapeutic interventions. The marine sponge glycolipid alpha-galactosylceramide (alphaGalCer) is the proto-typic NKT cell agonist, which elicits this function when bound to CD1d. However, our understanding of the in vivo properties of NKT cell agonists and the host factors that control their bioactivity remains very limited. In this report, we isolated the enzyme fatty acid amide hydrolase (FAAH) from mouse serum as an alphaGalCer-binding protein that modulates the induction of key effector functions of NKT cells in vivo. FAAH bound alphaGalCer in vivo and in vitro and was required for the efficient targeting of lipid antigens for CD1d presentation. Immunization of Faah-deficient mice with alphaGalCer resulted in a reduced systemic cytokine production, but enhanced expansion of splenic NKT cells. This distinct NKT response conferred a drastically increased adjuvant effect and strongly promoted protective CTL responses. Thus, our findings identify not only the presence of FAAH in normal mouse serum, but also its critical role in the tuning of immune responses to lipid antigens by orchestrating their transport and targeting for NKT cell activation. Our results suggest that the serum transport of lipid antigens directly shapes the quality of NKT cell responses, which could potentially be modulated in support of novel vaccination strategies.
Collapse
Affiliation(s)
- Stefan Freigang
- Department of Immunology and
Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California, USA.
Department of Chemistry, Brigham Young University, Provo, Utah, USA.
Committee on Immunology, University of Chicago, Chicago, Illinois, USA.
Howard Hughes Medical Institute, Chevy Chase, Maryland, USA
| | - Victoria Zadorozhny
- Department of Immunology and
Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California, USA.
Department of Chemistry, Brigham Young University, Provo, Utah, USA.
Committee on Immunology, University of Chicago, Chicago, Illinois, USA.
Howard Hughes Medical Institute, Chevy Chase, Maryland, USA
| | - Michele K. McKinney
- Department of Immunology and
Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California, USA.
Department of Chemistry, Brigham Young University, Provo, Utah, USA.
Committee on Immunology, University of Chicago, Chicago, Illinois, USA.
Howard Hughes Medical Institute, Chevy Chase, Maryland, USA
| | - Philippe Krebs
- Department of Immunology and
Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California, USA.
Department of Chemistry, Brigham Young University, Provo, Utah, USA.
Committee on Immunology, University of Chicago, Chicago, Illinois, USA.
Howard Hughes Medical Institute, Chevy Chase, Maryland, USA
| | - Rana Herro
- Department of Immunology and
Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California, USA.
Department of Chemistry, Brigham Young University, Provo, Utah, USA.
Committee on Immunology, University of Chicago, Chicago, Illinois, USA.
Howard Hughes Medical Institute, Chevy Chase, Maryland, USA
| | - Joanna Pawlak
- Department of Immunology and
Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California, USA.
Department of Chemistry, Brigham Young University, Provo, Utah, USA.
Committee on Immunology, University of Chicago, Chicago, Illinois, USA.
Howard Hughes Medical Institute, Chevy Chase, Maryland, USA
| | - Lisa Kain
- Department of Immunology and
Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California, USA.
Department of Chemistry, Brigham Young University, Provo, Utah, USA.
Committee on Immunology, University of Chicago, Chicago, Illinois, USA.
Howard Hughes Medical Institute, Chevy Chase, Maryland, USA
| | - Nicolas Schrantz
- Department of Immunology and
Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California, USA.
Department of Chemistry, Brigham Young University, Provo, Utah, USA.
Committee on Immunology, University of Chicago, Chicago, Illinois, USA.
Howard Hughes Medical Institute, Chevy Chase, Maryland, USA
| | - Kim Masuda
- Department of Immunology and
Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California, USA.
Department of Chemistry, Brigham Young University, Provo, Utah, USA.
Committee on Immunology, University of Chicago, Chicago, Illinois, USA.
Howard Hughes Medical Institute, Chevy Chase, Maryland, USA
| | - Yang Liu
- Department of Immunology and
Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California, USA.
Department of Chemistry, Brigham Young University, Provo, Utah, USA.
Committee on Immunology, University of Chicago, Chicago, Illinois, USA.
Howard Hughes Medical Institute, Chevy Chase, Maryland, USA
| | - Paul B. Savage
- Department of Immunology and
Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California, USA.
Department of Chemistry, Brigham Young University, Provo, Utah, USA.
Committee on Immunology, University of Chicago, Chicago, Illinois, USA.
Howard Hughes Medical Institute, Chevy Chase, Maryland, USA
| | - Albert Bendelac
- Department of Immunology and
Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California, USA.
Department of Chemistry, Brigham Young University, Provo, Utah, USA.
Committee on Immunology, University of Chicago, Chicago, Illinois, USA.
Howard Hughes Medical Institute, Chevy Chase, Maryland, USA
| | - Benjamin F. Cravatt
- Department of Immunology and
Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California, USA.
Department of Chemistry, Brigham Young University, Provo, Utah, USA.
Committee on Immunology, University of Chicago, Chicago, Illinois, USA.
Howard Hughes Medical Institute, Chevy Chase, Maryland, USA
| | - Luc Teyton
- Department of Immunology and
Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California, USA.
Department of Chemistry, Brigham Young University, Provo, Utah, USA.
Committee on Immunology, University of Chicago, Chicago, Illinois, USA.
Howard Hughes Medical Institute, Chevy Chase, Maryland, USA
| |
Collapse
|
182
|
Käsnänen H, Myllymäki MJ, Minkkilä A, Kataja AO, Saario SM, Nevalainen T, Koskinen AMP, Poso A. 3-Heterocycle-phenyl N-alkylcarbamates as FAAH inhibitors: design, synthesis and 3D-QSAR studies. ChemMedChem 2010; 5:213-31. [PMID: 20024981 DOI: 10.1002/cmdc.200900390] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Carbamates are a well-established class of fatty acid amide hydrolase (FAAH) inhibitors. Here we describe the synthesis of meta-substituted phenolic N-alkyl/aryl carbamates and their in vitro FAAH inhibitory activities. The most potent compound, 3-(oxazol-2yl)phenyl cyclohexylcarbamate (2 a), inhibited FAAH with a sub-nanomolar IC(50) value (IC(50)=0.74 nM). Additionally, we developed and validated three-dimensional quantitative structure-activity relationships (QSAR) models of FAAH inhibition combining the newly disclosed carbamates with our previously published inhibitors to give a total set of 99 compounds. Prior to 3D-QSAR modeling, the degree of correlation between FAAH inhibition and in silico reactivity was also established. Both 3D-QSAR methods used, CoMSIA and GRID/GOLPE, produced statistically significant models with coefficient of correlation for external prediction (R(2) (PRED)) values of 0.732 and 0.760, respectively. These models could be of high value in further FAAH inhibitor design.
Collapse
Affiliation(s)
- Heikki Käsnänen
- Department of Pharmaceutical Chemistry, University of Kuopio, P.O.Box 1627, 70211 Kuopio, Finland
| | | | | | | | | | | | | | | |
Collapse
|
183
|
Lovering AL, Lin LYC, Sewell EW, Spreter T, Brown ED, Strynadka NCJ. Structure of the bacterial teichoic acid polymerase TagF provides insights into membrane association and catalysis. Nat Struct Mol Biol 2010; 17:582-9. [PMID: 20400947 DOI: 10.1038/nsmb.1819] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2009] [Accepted: 03/02/2010] [Indexed: 12/23/2022]
Abstract
Teichoic acid polymers are composed of polyol-phosphate units and form a major component of Gram-positive bacterial cell walls. These anionic compounds perform a multitude of important roles in bacteria and are synthesized by monotopic membrane proteins of the TagF polymerase family. We have determined the structure of Staphylococcus epidermidis TagF to 2.7-A resolution from a construct that includes both the membrane-targeting region and the glycerol-phosphate polymerase domains. TagF possesses a helical region for interaction with the lipid bilayer, placing the active site at a suitable distance for access to the membrane-bound substrate. Characterization of active-site residue variants and analysis of a CDP-glycerol substrate complex suggest a mechanism for polymer synthesis. With the importance of teichoic acid in Gram-positive physiology, this elucidation of the molecular details of TagF function provides a critical new target in the development of novel anti-infectives.
Collapse
|
184
|
Abstract
Genome sequencing projects have uncovered thousands of uncharacterized enzymes in eukaryotic and prokaryotic organisms. Deciphering the physiological functions of enzymes requires tools to profile and perturb their activities in native biological systems. Activity-based protein profiling has emerged as a powerful chemoproteomic strategy to achieve these objectives through the use of chemical probes that target large swaths of enzymes that share active-site features. Here, we review activity-based protein profiling and its implementation to annotate the enzymatic proteome, with particular attention given to probes that target serine hydrolases, a diverse superfamily of enzymes replete with many uncharacterized members.
Collapse
Affiliation(s)
- Gabriel M. Simon
- From the Department of Chemical Physiology and Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, California 92037
| | - Benjamin F. Cravatt
- From the Department of Chemical Physiology and Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, California 92037
| |
Collapse
|
185
|
Labar G, Bauvois C, Borel F, Ferrer JL, Wouters J, Lambert DM. Crystal structure of the human monoacylglycerol lipase, a key actor in endocannabinoid signaling. Chembiochem 2010; 11:218-27. [PMID: 19957260 DOI: 10.1002/cbic.200900621] [Citation(s) in RCA: 141] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
2-Arachidonoylglycerol plays a major role in endocannabinoid signaling, and is tightly regulated by the monoacylglycerol lipase (MAGL). Here we report the crystal structure of human MAGL. The protein crystallizes as a dimer, and despite structural homologies to haloperoxidases and esterases, it distinguishes itself by a wide and hydrophobic access to the catalytic site. An apolar helix covering the active site also gives structural insight into the amphitropic character of MAGL, and likely explains how MAGL interacts with membranes to recruit its substrate. Docking of 2-arachidonoylglycerol highlights a hydrophobic and a hydrophilic cavity that accommodate the lipid into the catalytic site. Moreover, we identified Cys201 as the crucial residue in MAGL inhibition by N-arachidonylmaleimide, a sulfhydryl-reactive compound. Beside the advance in the knowledge of endocannabinoids degradation routes, the structure of MAGL paves the way for future medicinal chemistry works aimed at the design of new drugs exploiting 2-arachidonoylglycerol transmission.
Collapse
Affiliation(s)
- Geoffray Labar
- Unité de Chimie Pharmaceutique et de Radiopharmacie (CMFA), Louvain Drug Research Institute, Université catholique de Louvain, Faculté de Médecine, Avenue E. Mounier 73.40, 1200 Brussels, Belgium
| | | | | | | | | | | |
Collapse
|
186
|
Banni S, Di Marzo V. Effect of dietary fat on endocannabinoids and related mediators: consequences on energy homeostasis, inflammation and mood. Mol Nutr Food Res 2010; 54:82-92. [PMID: 20013888 DOI: 10.1002/mnfr.200900516] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Among the several known fatty acid-derived chemical signals, the endogenous ligands of cannabinoid receptors type-1 and -2, two G-protein-coupled receptors involved in several aspects of mammalian physiology and pathology, are perhaps those the levels of which have proven to be most sensitive to the fatty acid composition of the diet. The two most studied such ligands, known as endocannabinoids, are N-arachidonoyl-ethanolamine and 2-archidonoylglycerol, and are found in tissues together with other N-acyl-ethanolamines and 2-acylglycerols, not all of which activate the cannabinoid receptors, although several of them do exhibit important pharmacological effects. In this review article, we describe literature data indicating that the tissue concentrations of the endocannabinoids and related signalling molecules, and hence the activity of the respective receptors, can be modulated by modifying the fatty acid composition of the diet, and particularly its content in long chain PUFAs or in long chain PUFA precursors. We also discuss the potential impact of these diet-induced changes of endocannabinoid tone on three of the major pathological conditions in which cannabinoid receptors have been involved, that is metabolic dysfunctions, inflammation and affective disorders.
Collapse
Affiliation(s)
- Sebastiano Banni
- Dipartimento di Biologia Sperimentale, Università di Cagliari, Cagliari, Italy
| | | |
Collapse
|
187
|
Methylation and acetylation of 15-hydroxyanandamide modulate its interaction with the endocannabinoid system. Biochimie 2010; 92:378-87. [DOI: 10.1016/j.biochi.2010.01.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2009] [Accepted: 01/03/2010] [Indexed: 11/23/2022]
|
188
|
Long JZ, Jin X, Adibekian A, Li W, Cravatt BF. Characterization of tunable piperidine and piperazine carbamates as inhibitors of endocannabinoid hydrolases. J Med Chem 2010; 53:1830-42. [PMID: 20099888 DOI: 10.1021/jm9016976] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Monoacylglycerol lipase (MAGL) and fatty acid amide hydrolase (FAAH) are two enzymes from the serine hydrolase superfamily that degrade the endocannabinoids 2-arachidonoylglycerol and anandamide, respectively. We have recently discovered that MAGL and FAAH are both inhibited by carbamates bearing an N-piperidine/piperazine group. Piperidine/piperazine carbamates show excellent in vivo activity, raising brain endocannabinoid levels and producing CB1-dependent behavioral effects in mice, suggesting that they represent a promising class of inhibitors for studying the endogenous functions of MAGL and FAAH. Herein, we disclose a full account of the syntheses, structure-activity relationships, and inhibitory activities of piperidine/piperazine carbamates against members of the serine hydrolase family. These scaffolds can be tuned for MAGL-selective or dual MAGL-FAAH inhibition by the attachment of an appropriately substituted bisarylcarbinol or aryloxybenzyl moiety, respectively, on the piperidine/piperazine ring. Modifications to the piperidine/piperazine ring ablated inhibitory activity, suggesting a strict requirement for a six-membered ring to maintain potency.
Collapse
Affiliation(s)
- Jonathan Z Long
- The Skaggs Institute for Chemical Biology and Department of Chemical Physiology, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, California 92037, USA
| | | | | | | | | |
Collapse
|
189
|
Gattinoni S, De Simone C, Dallavalle S, Fezza F, Nannei R, Amadio D, Minetti P, Quattrociocchi G, Caprioli A, Borsini F, Cabri W, Penco S, Merlini L, Maccarrone M. Enol Carbamates as Inhibitors of Fatty Acid Amide Hydrolase (FAAH) Endowed with High Selectivity for FAAH over the Other Targets of the Endocannabinoid System. ChemMedChem 2010; 5:357-60. [DOI: 10.1002/cmdc.200900472] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
190
|
Ueda N, Tsuboi K, Uyama T. N-acylethanolamine metabolism with special reference to N-acylethanolamine-hydrolyzing acid amidase (NAAA). Prog Lipid Res 2010; 49:299-315. [PMID: 20152858 DOI: 10.1016/j.plipres.2010.02.003] [Citation(s) in RCA: 124] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
N-acylethanolamines (NAEs) constitute a class of bioactive lipid molecules present in animal and plant tissues. Among the NAEs, N-arachidonoylethanolamine (anandamide), N-palmitoylethanolamine, and N-oleoylethanolamine attract much attention due to cannabimimetic activity as an endocannabinoid, anti-inflammatory and analgesic activities, and anorexic activity, respectively. In mammalian tissues, NAEs are formed from glycerophospholipids through the phosphodiesterase-transacylation pathway consisting of Ca(2+)-dependent N-acyltransferase and N-acylphosphatidylethanolamine-hydrolyzing phospholipase D. Recent studies revealed the presence of alternative pathways and enzymes responsible for the NAE formation. As for the degradation of NAEs, fatty acid amide hydrolase (FAAH), which hydrolyzes NAEs to fatty acids and ethanolamine, plays a central role. However, a lysosomal enzyme referred to as NAE-hydrolyzing acid amidase (NAAA) also catalyzes the same reaction and may be a new target for the development of therapeutic drugs. In this article we discuss recent progress in the studies on the enzymes involved in the biosynthesis and degradation of NAEs with special reference to NAAA.
Collapse
Affiliation(s)
- Natsuo Ueda
- Department of Biochemistry, Kagawa University School of Medicine, 1750-1 Ikenobe, Miki, Kagawa, Japan
| | | | | |
Collapse
|
191
|
Biochemical characterization, membrane association and identification of amino acids essential for the function of Alg11 from Saccharomyces cerevisiae, an alpha1,2-mannosyltransferase catalysing two sequential glycosylation steps in the formation of the lipid-linked core oligosaccharide. Biochem J 2010; 426:205-17. [PMID: 19929855 DOI: 10.1042/bj20091121] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The biosynthesis of asparagine-linked glycans occurs in an evolutionarily conserved manner with the assembly of the unique lipid-linked oligosaccharide precursor Glc3Man9GlcNAc2-PP-Dol at the ER (endoplasmic reticulum). In the present study we characterize Alg11 from yeast as a mannosyltransferase catalysing the sequential transfer of two alpha1,2-linked mannose residues from GDP-mannose to Man3GlcNAc2-PP-Dol and subsequently to Man4GlcNAc2-PP-Dol forming the Man5GlcNAc2-PP-Dol intermediate at the cytosolic side of the ER before flipping to the luminal side. Alg11 is predicted to contain three hydrophobic transmembrane-spanning helices. Using Alg11 topology reporter fusion constructs, we show that only the N-terminal domain fulfils this criterion. Surprisingly, this domain can be deleted without disturbing glycosyltransferase function and membrane association, indicating also that the other two hydrophobic domains contribute to ER localization, but in a non-transmembrane manner. By site-directed mutagenesis we investigated amino acids important for transferase activity. We demonstrate that the first glutamate residue in the EX7E motif, conserved in a variety of glycosyltransferases, is more critical than the second, and loss of Alg11 function occurs only when both glutamate residues are exchanged, or when the mutation of the first glutamate residue is combined with replacement of another amino acid in the motif. This indicates that perturbations in EX7E are not restricted to the second glutamate residue. Moreover, Gly85 and Gly87, within a glycine-rich domain as part of a potential flexible loop, were found to be required for Alg11 function. Similarly, a conserved lysine residue, Lys319, was identified as being important for activity, which could be involved in the binding of the phosphate of the glycosyl donor.
Collapse
|
192
|
Acevedo O, Jorgensen WL. Advances in quantum and molecular mechanical (QM/MM) simulations for organic and enzymatic reactions. Acc Chem Res 2010; 43:142-51. [PMID: 19728702 DOI: 10.1021/ar900171c] [Citation(s) in RCA: 186] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Application of combined quantum and molecular mechanical (QM/MM) methods focuses on predicting activation barriers and the structures of stationary points for organic and enzymatic reactions. Characterization of the factors that stabilize transition structures in solution and in enzyme active sites provides a basis for design and optimization of catalysts. Continued technological advances allowed for expansion from prototypical cases to mechanistic studies featuring detailed enzyme and condensed-phase environments with full integration of the QM calculations and configurational sampling. This required improved algorithms featuring fast QM methods, advances in computing changes in free energies including free-energy perturbation (FEP) calculations, and enhanced configurational sampling. In particular, the present Account highlights development of the PDDG/PM3 semi-empirical QM method, computation of multi-dimensional potentials of mean force (PMF), incorporation of on-the-fly QM in Monte Carlo (MC) simulations, and a polynomial quadrature method for efficient modeling of proton-transfer reactions. The utility of this QM/MM/MC/FEP methodology is illustrated for a variety of organic reactions including substitution, decarboxylation, elimination, and pericyclic reactions. A comparison to experimental kinetic results on medium effects has verified the accuracy of the QM/MM approach in the full range of solvents from hydrocarbons to water to ionic liquids. Corresponding results from ab initio and density functional theory (DFT) methods with continuum-based treatments of solvation reveal deficiencies, particularly for protic solvents. Also summarized in this Account are three specific QM/MM applications to biomolecular systems: (1) a recent study that clarified the mechanism for the reaction of 2-pyrone derivatives catalyzed by macrophomate synthase as a tandem Michael-aldol sequence rather than a Diels-Alder reaction, (2) elucidation of the mechanism of action of fatty acid amide hydrolase (FAAH), an unusual Ser-Ser-Lys proteolytic enzyme, and (3) the construction of enzymes for Kemp elimination of 5-nitrobenzisoxazole that highlights the utility of QM/MM in the design of artificial enzymes.
Collapse
Affiliation(s)
- Orlando Acevedo
- Department of Chemistry and Biochemistry, Auburn University, Auburn, Alabama 36849
| | - William L. Jorgensen
- Department of Chemistry, Yale University, 225 Prospect Street, New Haven, Connecticut 06520-8107
| |
Collapse
|
193
|
Mileni M, Garfunkle J, Ezzili C, Kimball FS, Cravatt BF, Stevens RC, Boger DL. X-ray crystallographic analysis of alpha-ketoheterocycle inhibitors bound to a humanized variant of fatty acid amide hydrolase. J Med Chem 2010; 53:230-40. [PMID: 19924997 PMCID: PMC2804032 DOI: 10.1021/jm9012196] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Three cocrystal X-ray structures of the alpha-ketoheterocycle inhibitors 3-5 bound to a humanized variant of fatty acid amide hydrolase (FAAH) are disclosed and comparatively discussed alongside those of 1 (OL-135) and its isomer 2. These five X-ray structures systematically probe each of the three active site regions key to substrate or inhibitor binding: (1) the conformationally mobile acyl chain-binding pocket and membrane access channel responsible for fatty acid amide substrate and inhibitor acyl chain binding, (2) the atypical active site catalytic residues and surrounding oxyanion hole that covalently binds the core of the alpha-ketoheterocycle inhibitors captured as deprotonated hemiketals mimicking the tetrahedral intermediate of the enzyme-catalyzed reaction, and (3) the cytosolic port and its uniquely important imbedded ordered water molecules and a newly identified anion binding site. The detailed analysis of their key active site interactions and their implications on the interpretation of the available structure-activity relationships are discussed providing important insights for future design.
Collapse
Affiliation(s)
- Mauro Mileni
- Department of Molecular Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037
| | - Joie Garfunkle
- Department of Chemistry, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037
- The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037
| | - Cyrine Ezzili
- Department of Chemistry, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037
- The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037
| | - F. Scott Kimball
- Department of Chemistry, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037
- The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037
| | - Benjamin F. Cravatt
- Department of Chemical Physiology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037
- The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037
| | - Raymond C. Stevens
- Department of Chemistry, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037
- Department of Molecular Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037
| | - Dale L. Boger
- Department of Chemistry, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037
- The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037
| |
Collapse
|
194
|
Computing Free-Energy Profiles Using Multidimensional Potentials of Mean Force and Polynomial Quadrature Methods. ACTA ACUST UNITED AC 2010. [DOI: 10.1016/s1574-1400(10)06003-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
|
195
|
Ohtaki A, Murata K, Sato Y, Noguchi K, Miyatake H, Dohmae N, Yamada K, Yohda M, Odaka M. Structure and characterization of amidase from Rhodococcus sp. N-771: Insight into the molecular mechanism of substrate recognition. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2010; 1804:184-92. [DOI: 10.1016/j.bbapap.2009.10.001] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2009] [Revised: 09/27/2009] [Accepted: 10/01/2009] [Indexed: 11/26/2022]
|
196
|
Alapafuja SO, Nikas SP, Shukla VG, Papanastasiou I, Makriyannis A. Microwave Assisted Synthesis of Sodium Sulfonates Precursors of Sulfonyl Chlorides and Fluorides. Tetrahedron Lett 2009; 50:7028-7031. [PMID: 21132105 PMCID: PMC2994267 DOI: 10.1016/j.tetlet.2009.09.167] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
We describe the use of a microwave reaction for the conversion of various bromides to sodium sulfonates that have been further elaborated to sulfonyl chlorides. This new approach leads to much improved yields and shorter reaction times. Representative sulfonyl chlorides serve as precursors for the respective sulfonyl fluorides that are potent inhibitors of the fatty acid amide hydrolase.
Collapse
Affiliation(s)
- Shakiru O. Alapafuja
- Center for Drug Discovery, Northeastern University, 116 Mugar Life Sciences Building, 360 Huntington Avenue, Boston, MA, 02115, USA
| | - Spyros P. Nikas
- Center for Drug Discovery, Northeastern University, 116 Mugar Life Sciences Building, 360 Huntington Avenue, Boston, MA, 02115, USA
| | - Vidyanand G. Shukla
- Center for Drug Discovery, Northeastern University, 116 Mugar Life Sciences Building, 360 Huntington Avenue, Boston, MA, 02115, USA
| | - Ioannis Papanastasiou
- Center for Drug Discovery, Northeastern University, 116 Mugar Life Sciences Building, 360 Huntington Avenue, Boston, MA, 02115, USA
| | - Alexandros Makriyannis
- Center for Drug Discovery, Northeastern University, 116 Mugar Life Sciences Building, 360 Huntington Avenue, Boston, MA, 02115, USA
| |
Collapse
|
197
|
Bertrand T, Augé F, Houtmann J, Rak A, Vallée F, Mikol V, Berne PF, Michot N, Cheuret D, Hoornaert C, Mathieu M. Structural basis for human monoglyceride lipase inhibition. J Mol Biol 2009; 396:663-73. [PMID: 19962385 DOI: 10.1016/j.jmb.2009.11.060] [Citation(s) in RCA: 133] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2009] [Revised: 11/24/2009] [Accepted: 11/24/2009] [Indexed: 11/16/2022]
Abstract
Monoglyceride lipase (MGL) is a serine hydrolase that hydrolyses 2-arachidonoylglycerol (2-AG) into arachidonic acid and glycerol. 2-AG is an endogenous ligand of cannabinoid receptors, involved in various physiological processes in the brain. We present here the first crystal structure of human MGL in its apo form and in complex with the covalent inhibitor SAR629. MGL shares the classic fold of the alpha/beta hydrolase family but depicts an unusually large hydrophobic occluded tunnel with a highly flexible lid at its entry and the catalytic triad buried at its end. Structures reveal the configuration of the catalytic triad and the shape and nature of the binding site of 2-AG. The bound structure of SAR629 highlights the key interactions for productive binding with MGL. The shape of the tunnel suggests a high druggability of the protein and provides an attractive template for drug discovery.
Collapse
Affiliation(s)
- T Bertrand
- Department of Structural Biology, Sanofi-Aventis, 13 Quai Jules Guesde, 94403 Vitry-sur-Seine cedex, France.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
198
|
Reggio PH. Toward the design of cannabinoid CB1 receptor inverse agonists and neutral antagonists. Drug Dev Res 2009. [DOI: 10.1002/ddr.20337] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
199
|
Zhao YS, Zheng QC, Zhang HX, Chu HY, Sun CC. Homology modelling and molecular dynamics study of human fatty acid amide hydrolase. MOLECULAR SIMULATION 2009. [DOI: 10.1080/08927020903033133] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
200
|
Morera E, De Petrocellis L, Morera L, Moriello AS, Ligresti A, Nalli M, Woodward DF, Di Marzo V, Ortar G. Synthesis and biological evaluation of piperazinyl carbamates and ureas as fatty acid amide hydrolase (FAAH) and transient receptor potential (TRP) channel dual ligands. Bioorg Med Chem Lett 2009; 19:6806-9. [DOI: 10.1016/j.bmcl.2009.09.033] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2009] [Revised: 09/09/2009] [Accepted: 09/10/2009] [Indexed: 12/28/2022]
|