Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 98(16): 9371-6, 2001 Jul 31.
Article in English | MEDLINE | ID: mdl-11470906

ABSTRACT

The medicinal properties of marijuana have been recognized for centuries, but clinical and societal acceptance of this drug of abuse as a potential therapeutic agent remains fiercely debated. An attractive alternative to marijuana-based therapeutics would be to target the molecular pathways that mediate the effects of this drug. To date, these neural signaling pathways have been shown to comprise a cannabinoid receptor (CB(1)) that binds the active constituent of marijuana, tetrahydrocannabinol (THC), and a postulated endogenous CB(1) ligand anandamide. Although anandamide binds and activates the CB(1) receptor in vitro, this compound induces only weak and transient cannabinoid behavioral effects in vivo, possibly a result of its rapid catabolism. Here we show that mice lacking the enzyme fatty acid amide hydrolase (FAAH(-/-)) are severely impaired in their ability to degrade anandamide and when treated with this compound, exhibit an array of intense CB(1)-dependent behavioral responses, including hypomotility, analgesia, catalepsy, and hypothermia. FAAH(-/-)-mice possess 15-fold augmented endogenous brain levels of anandamide and display reduced pain sensation that is reversed by the CB(1) antagonist SR141716A. Collectively, these results indicate that FAAH is a key regulator of anandamide signaling in vivo, setting an endogenous cannabinoid tone that modulates pain perception. FAAH may therefore represent an attractive pharmaceutical target for the treatment of pain and neuropsychiatric disorders.


Subject(s)
Amidohydrolases/physiology , Arachidonic Acids/pharmacology , Behavior, Animal/drug effects , Cannabinoids/metabolism , Signal Transduction , Amidohydrolases/genetics , Animals , Brain/metabolism , Endocannabinoids , Ethanolamines/metabolism , Mice , Mice, Knockout , Pain Threshold , Polyunsaturated Alkamides , Receptors, Cannabinoid , Receptors, Drug/metabolism
2.
Vitam Horm ; 62: 95-131, 2001.
Article in English | MEDLINE | ID: mdl-11345902

ABSTRACT

Fatty acid amides (FAAs) represent a growing family of biologically active lipids implicated in a diverse range of cellular and physiological processes. At present, two general types of fatty acid amides, the N-acylethanolamines (NAEs) and the fatty acid primary amides (FAPAs), have been identified as potential physiological neuromodulators/neurotransmitters in mammals. Representative members of these two subfamilies include the endocannabinoid NAE anandamide and the sleep-inducing FAPA oleamide. In this Chapter, molecular mechanisms proposed for the biosynthesis and inactivation of FAAs are critically evaluated, with an emphasis placed on the biochemical and cell biological properties of proteins thought to mediate these processes.


Subject(s)
Amides/metabolism , Fatty Acids/physiology , Neurotransmitter Agents/physiology , Amidohydrolases/metabolism , Animals , Biological Transport , Cannabinoid Receptor Modulators , Ethanolamines/metabolism , Fatty Acids/metabolism , Humans , Neurotransmitter Agents/metabolism
3.
Biochemistry ; 40(20): 6107-15, 2001 May 22.
Article in English | MEDLINE | ID: mdl-11352748

ABSTRACT

Fatty acid amide hydrolase (FAAH) is a mammalian integral membrane enzyme that catabolizes several neuromodulatory fatty acid amides, including the endogenous cannabinoid anandamide and the sleep-inducing lipid oleamide. FAAH belongs to a large group of hydrolytic enzymes termed the amidase signature (AS) family that is defined by a conserved, linear AS sequence of approximately 130 amino acids. Members of the AS family display strikingly different substrate selectivities, yet the primary structural regions responsible for defining substrate recognition in these enzymes remain unknown. In this study, a series of unbranched p-nitroanilide (pNA) substrates ranging from 6 to 20 carbons in length was used to probe the acyl chain binding specificity of FAAH, revealing that this enzyme exhibits a strong preference for acyl chains 9 carbons in length or longer. A fluorophosphonate inhibitor of FAAH containing a photoactivatable benzophenone group was synthesized and used to locate a region of the enzyme implicated in substrate binding. Protease digestion and mass spectrometry analysis of FAAH-inhibitor conjugates identified the major site of cross-linking as residues 487-493. Site-directed mutagenesis revealed that a single residue in this region, I491, strongly influenced substrate specificity of FAAH. For example, an I491A mutant displayed a greatly reduced binding affinity for medium-chain pNA substrates (7-12 carbons) but maintained nearly wild-type binding and catalytic constants for longer chain substrates (14-20 carbons). Mutation of I491 to aromatic or more polar residues generated enzymes with relative hydrolytic efficiencies for medium- versus long-chain pNAs that varied up to 90-fold. Collectively, these studies indicate that I491 participates in hydrophobic binding interactions with medium-chain FAAH substrates. Additionally, the significant changes in substrate selectivity achieved by single amino acid changes suggest that FAAH possesses a rather malleable substrate binding domain and may serve, along with other AS enzymes, as a template for the engineering of amidases with novel and/or tailored specificities.


Subject(s)
Amidohydrolases/chemistry , Amidohydrolases/metabolism , Fatty Acids/chemistry , Fatty Acids/metabolism , Alanine/genetics , Amidohydrolases/antagonists & inhibitors , Amidohydrolases/genetics , Amino Acid Sequence , Binding Sites/genetics , Binding, Competitive/genetics , Catalysis , Chromatography, High Pressure Liquid , Cross-Linking Reagents/chemistry , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/metabolism , Isoleucine/genetics , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Structure, Tertiary/genetics , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Substrate Specificity/genetics
4.
Proteomics ; 1(9): 1067-71, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11990500

ABSTRACT

The field of biochemistry is currently faced with the enormous challenge of assigning functional significance to more than thirty thousand predicted protein products encoded by the human genome. In order to accomplish this daunting task, methods will be required that facilitate the global analysis of proteins in complex biological systems. Recently, methods have been described for simultaneously monitoring the activity of multiple enzymes in crude proteomes based on their reactivity with tagged chemical probes. These activity based probes (ABPs) have used either radiochemical or biotin/avidin-based detection methods to allow consolidated visualization of numerous enzyme activities. Here we report the synthesis and evaluation of fluorescent activity based probes for the serine hydrolase super-family of enzymes. The fluorescent methods detailed herein provide superior throughput, sensitivity, and quantitative accuracy when compared to previously described ABPs, and provide a straight-forward platform for high-throughput proteome analysis.


Subject(s)
Fluorescent Dyes/metabolism , Molecular Probe Techniques , Proteome , Serine Endopeptidases/metabolism , Serine/analogs & derivatives , Animals , COS Cells , Fluorescent Dyes/chemical synthesis , Humans , Molecular Structure , Rats
5.
Bioorg Med Chem Lett ; 10(23): 2613-6, 2000 Dec 04.
Article in English | MEDLINE | ID: mdl-11128635

ABSTRACT

Fatty acid amide hydrolase (FAAH), also referred to as oleamide hydrolase and anandamide amidohydrolase, is a serine hydrolase responsible for the degradation of endogenous oleamide and anandamide, fatty acid amides that function as chemical messengers. FAAH hydrolyzes a range of fatty acid amides, and the present study examines the relative rates of hydrolysis of a variety of natural and unnatural fatty acid primary amide substrates using pure recombinant rat FAAH.


Subject(s)
Amidohydrolases/metabolism , Animals , Hydrolysis , Rats , Recombinant Proteins/metabolism , Substrate Specificity
6.
Proc Natl Acad Sci U S A ; 97(10): 5044-9, 2000 May 09.
Article in English | MEDLINE | ID: mdl-10805767

ABSTRACT

The development of exceptionally potent inhibitors of fatty acid amide hydrolase (FAAH), the enzyme responsible for the degradation of oleamide (an endogenous sleep-inducing lipid), and anandamide (an endogenous ligand for cannabinoid receptors) is detailed. The inhibitors may serve as useful tools to clarify the role of endogenous oleamide and anandamide and may prove to be useful therapeutic agents for the treatment of sleep disorders or pain. The combination of several features-an optimal C12-C8 chain length, pi-unsaturation introduction at the corresponding arachidonoyl Delta(8,9)/Delta(11,12) and oleoyl Delta(9,10) location, and an alpha-keto N4 oxazolopyridine with incorporation of a second weakly basic nitrogen provided FAAH inhibitors with K(i)s that drop below 200 pM and are 10(2)-10(3) times more potent than the corresponding trifluoromethyl ketones.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Arachidonic Acids/pharmacokinetics , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Heterocyclic Compounds/chemistry , Heterocyclic Compounds/pharmacology , Oleic Acids/metabolism , Animals , COS Cells , Cannabinoids/pharmacokinetics , Cell Membrane/enzymology , Cerebrosides/metabolism , Drug Design , Endocannabinoids , Enzyme Inhibitors/chemical synthesis , Heterocyclic Compounds/chemical synthesis , Kinetics , Liver/enzymology , Polyunsaturated Alkamides , Rats , Recombinant Proteins/antagonists & inhibitors , Structure-Activity Relationship , Transfection
7.
J Biol Chem ; 275(25): 19177-84, 2000 Jun 23.
Article in English | MEDLINE | ID: mdl-10764768

ABSTRACT

Fatty acid amide hydrolase (FAAH) is a mammalian integral membrane enzyme responsible for the hydrolysis of a number of neuromodulatory fatty acid amides, including the endogenous cannabinoid anandamide and the sleep-inducing lipid oleamide. FAAH belongs to a large class of hydrolytic enzymes termed the "amidase signature family," whose members are defined by a conserved stretch of approximately 130 amino acids termed the "amidase signature sequence." Recently, site-directed mutagenesis studies of FAAH have targeted a limited number of conserved residues in the amidase signature sequence of the enzyme, identifying Ser-241 as the catalytic nucleophile and Lys-142 as an acid/base catalyst. The roles of several other conserved residues with potentially important and/or overlapping catalytic functions have not yet been examined. In this study, we have mutated all potentially catalytic residues in FAAH that are conserved among members of the amidase signature family, and have assessed their individual roles in catalysis through chemical labeling and kinetic methods. Several of these residues appear to serve primarily structural roles, as their mutation produced FAAH variants with considerable catalytic activity but reduced expression in prokaryotic and/or eukaryotic systems. In contrast, five mutations, K142A, S217A, S218A, S241A, and R243A, decreased the amidase activity of FAAH greater than 100-fold without detectably impacting the structural integrity of the enzyme. The pH rate profiles, amide/ester selectivities, and fluorophosphonate reactivities of these mutants revealed distinct catalytic roles for each residue. Of particular interest, one mutant, R243A, displayed uncompromised esterase activity but severely reduced amidase activity, indicating that the amidase and esterase efficiencies of FAAH can be functionally uncoupled. Collectively, these studies provide evidence that amidase signature enzymes represent a large class of serine-lysine catalytic dyad hydrolases whose evolutionary distribution rivals that of the catalytic triad superfamily.


Subject(s)
Amidohydrolases/metabolism , Conserved Sequence , Amidohydrolases/chemistry , Amino Acid Sequence , Catalysis , Hydrogen-Ion Concentration , Kinetics , Molecular Sequence Data , Sequence Homology, Amino Acid
8.
Proc Natl Acad Sci U S A ; 96(26): 14694-9, 1999 Dec 21.
Article in English | MEDLINE | ID: mdl-10611275

ABSTRACT

With the postgenome era rapidly approaching, new strategies for the functional analysis of proteins are needed. To date, proteomics efforts have primarily been confined to recording variations in protein level rather than activity. The ability to profile classes of proteins on the basis of changes in their activity would greatly accelerate both the assignment of protein function and the identification of potential pharmaceutical targets. Here, we describe the chemical synthesis and utility of an active-site directed probe for visualizing dynamics in the expression and function of an entire enzyme family, the serine hydrolases. By reacting this probe, a biotinylated fluorophosphonate referred to as FP-biotin, with crude tissue extracts, we quickly and with high sensitivity detect numerous serine hydrolases, many of which display tissue-restricted patterns of expression. Additionally, we show that FP-biotin labels these proteins in an activity-dependent manner that can be followed kinetically, offering a powerful means to monitor dynamics simultaneously in both protein function and expression.


Subject(s)
Biotin/analogs & derivatives , Gene Expression Profiling/methods , Organophosphorus Compounds/pharmacology , Serine Endopeptidases/isolation & purification , Serine Proteinase Inhibitors/pharmacology , Animals , Biotin/chemical synthesis , Biotin/pharmacology , Brain/enzymology , Catalytic Domain/drug effects , Drug Design , Male , Organophosphorus Compounds/chemical synthesis , Rats , Serine Endopeptidases/drug effects , Serine Proteinase Inhibitors/chemical synthesis , Tissue Distribution
9.
Biochemistry ; 38(43): 14125-30, 1999 Oct 26.
Article in English | MEDLINE | ID: mdl-10571985

ABSTRACT

The greater reactivity of esters relative to amides has typically been reflected in their faster rates of both solvolysis and enzymatic hydrolysis. In contrast to this general principle, the serine hydrolytic enzyme fatty acid amide hydrolase (FAAH) was found to degrade amides and esters with equivalent catalytic efficiencies. Mutation of a single lysine residue (K142) to alanine (K142A) abolished this property, generating a catalytically compromised enzyme that hydrolyzed esters more than 500-fold faster than amides. Conversion of this same lysine residue to glutamic acid (K142E) produced an enzyme that also displayed severely diminished catalytic activity, but one that now maintained FAAH's ability to react with amides and esters at competitive rates. The significant catalytic defects exhibited by both the K142A and K142E mutants, in conjunction with their altered pH-rate profiles, support a role for lysine 142 as a general base involved in the activation of FAAH's serine nucleophile. Moreover, the dramatically different amide versus ester selectivities observed for the K142A and K142E mutants reveal that FAAH's catalytic efficiency and catalytic selectivity depend on distinguishable properties of the same residue, with the former relying on a strong catalytic base and the latter requiring coupled general acid-base catalysis. We hypothesize that FAAH's unusual catalytic properties may empower the enzyme to function effectively as both an amidase and esterase in vivo.


Subject(s)
Amides/metabolism , Amidohydrolases/metabolism , Acylation , Amides/chemistry , Amidohydrolases/chemistry , Amidohydrolases/genetics , Amino Acid Substitution , Animals , Binding, Competitive , Catalysis , Enzyme Activation , Esters , Hydrogen-Ion Concentration , Hydrolysis , Kinetics , Oleic Acids/metabolism , Point Mutation , Rats , Structure-Activity Relationship , Substrate Specificity
10.
Biochemistry ; 38(31): 9804-12, 1999 Aug 03.
Article in English | MEDLINE | ID: mdl-10433686

ABSTRACT

Fatty acid amide hydrolase (FAAH) is a membrane-bound enzyme responsible for the catabolism of neuromodulatory fatty acid amides, including anandamide and oleamide. FAAH's primary structure identifies this enzyme as a member of a diverse group of alkyl amidases, known collectively as the "amidase signature family". At present, this enzyme family's catalytic mechanism remains poorly understood. In this study, we investigated the catalytic features of FAAH through mutagenesis, affinity labeling, and steady-state kinetic methods. In particular, we focused on the respective roles of three serine residues that are conserved in all amidase signature enzymes (S217, S218, and S241 in FAAH). Mutation of each of these serines to alanine resulted in a FAAH enzyme bearing significant catalytic defects, with the S217A and S218A mutants showing 2300- and 95-fold reductions in k(cat), respectively, and the S241A mutant exhibiting no detectable catalytic activity. The double S217A:S218A FAAH mutant displayed a 230 000-fold decrease in k(cat), supporting independent catalytic functions for these serine residues. Affinity labeling of FAAH with a specific nucleophile reactive inhibitor, ethoxy oleoyl fluorophosphonate, identified S241 as the enzyme's catalytic nucleophile. The pH dependence of FAAH's k(cat) and k(cat)/K(m) implicated a base involved in catalysis with a pK(a) of 7.9. Interestingly, mutation of each of FAAH's conserved histidines (H184, H358, and H449) generated active enzymes, indicating that FAAH does not contain a Ser-His-Asp catalytic triad commonly found in other mammalian serine hydrolytic enzymes. The unusual properties of FAAH identified here suggest that this enzyme, and possibly the amidase signature family as a whole, may hydrolyze amides by a novel catalytic mechanism.


Subject(s)
Amidohydrolases/chemistry , Amidohydrolases/genetics , Mutagenesis, Site-Directed , Affinity Labels/chemistry , Affinity Labels/metabolism , Amidohydrolases/antagonists & inhibitors , Amidohydrolases/metabolism , Amino Acid Sequence , Animals , Catalysis , Circular Dichroism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Humans , Kinetics , Mice , Molecular Sequence Data , Multigene Family , Oleic Acids/chemistry , Oleic Acids/metabolism , Rats , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Serine/genetics , Serine/metabolism
11.
Bioorg Med Chem Lett ; 9(2): 265-70, 1999 Jan 18.
Article in English | MEDLINE | ID: mdl-10021942

ABSTRACT

The examination of a series of trifluoromethyl ketone inhibitors of Fatty Acid Amide Hydrolase (FAAH, oleamide hydrolase, anandamide amidohydrolase) is detailed in efforts that define structural and conformational properties that contribute to enzyme inhibition and substrate binding. The results imply an extended bound conformation, highlight a role for the presence, position, and stereochemistry of a delta cis double bond, and suggest little apparent role for C11-C18/C22 of the fatty acid amide substrates.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Inhibitory Concentration 50 , Ketones/chemistry , Oleic Acids/chemistry , Temperature
12.
Biochemistry ; 37(43): 15177-87, 1998 Oct 27.
Article in English | MEDLINE | ID: mdl-9790682

ABSTRACT

Fatty acid amide hydrolase (FAAH) is an integral membrane protein responsible for the hydrolysis of a number of primary and secondary fatty acid amides, including the neuromodulatory compounds anandamide and oleamide. Analysis of FAAH's primary sequence reveals the presence of a single predicted transmembrane domain at the extreme N-terminus of the enzyme. A mutant form of the rat FAAH protein lacking this N-terminal transmembrane domain (DeltaTM-FAAH) was generated and, like wild type FAAH (WT-FAAH), was found to be tightly associated with membranes when expressed in COS-7 cells. Recombinant forms of WT- and DeltaTM-FAAH expressed and purified from Escherichia coli exhibited essentially identical enzymatic properties which were also similar to those of the native enzyme from rat liver. Analysis of the oligomerization states of WT- and DeltaTM-FAAH by chemical cross-linking, sedimentation velocity analytical ultracentrifugation, and size exclusion chromatography indicated that both enzymes were oligomeric when membrane-bound and after solubilization. However, WT-FAAH consistently behaved as a larger oligomer than DeltaTM-FAAH. Additionally, SDS-PAGE analysis of the recombinant proteins identified the presence of SDS-resistant oligomers for WT-FAAH, but not for DeltaTM-FAAH. Self-association through FAAH's transmembrane domain was further demonstrated by a FAAH transmembrane domain-GST fusion protein which formed SDS-resistant dimers and large oligomeric assemblies in solution.


Subject(s)
Amidohydrolases/chemistry , Membrane Proteins/chemistry , Peptide Fragments/genetics , Sequence Deletion , Amidohydrolases/genetics , Amidohydrolases/metabolism , Animals , COS Cells , Escherichia coli/genetics , Glutathione Transferase/genetics , Humans , Liver/enzymology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Structure, Tertiary , Rats , Recombinant Fusion Proteins/chemistry , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification
13.
Bioorg Med Chem Lett ; 8(6): 613-8, 1998 Mar 17.
Article in English | MEDLINE | ID: mdl-9871570

ABSTRACT

2-Octyl gamma-bromoacetoacetate (O gamma Br), an endogenous compound originally isolated from human cerebrospinal fluid (CSF), has previously been demonstrated to increase REM sleep duration in cats. Based on the chemical structure of O gamma Br and its reported sleep-inducing effects, we synthesized O gamma Br along with chemically related analogs and tested these compounds as inhibitors of fatty acid amide hydrolase (FAAH), a brain enzyme that degrades neuromodulatory fatty acid amides. O gamma Br was found to competitively inhibit FAAH activity with IC50 and Ki values of 2.6 microM and 0.8 microM, respectively [for the (R)-enantiomer of O gamma Br (1)]. A set of synthetic analogs of O gamma Br was examined to define the structural features required for FAAH inhibition and inhibitor potencies were assessed at pH 9.0 (near the pH optimum of FAAH) and pH 7.0. Interestingly, at pH 7.0 the gamma-halo beta-keto ester inhibitors proved to be significantly more potent than the trifluoromethyl ketone of oleic acid, one of the most potent FAAH inhibitors described to date. This study supports the possibility that O gamma Br may be a physiological regulator of FAAH activity and fatty acid amide levels in vivo. Additionally, the characterization of gamma-halo beta-keto esters as powerful FAAH inhibitors near physiological pH may aid in future studies of the enzymology and biological properties of FAAH.


Subject(s)
Acetoacetates/pharmacology , Amidohydrolases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Sleep/drug effects , Animals , Binding, Competitive , Cats , Humans , Hydrogen-Ion Concentration , Models, Chemical
SELECTION OF CITATIONS
SEARCH DETAIL
...