FA2H

14,000,000 Leading Edge Experts on the ideXlab platform

Scan Science and Technology

Contact Leading Edge Experts & Companies

Scan Science and Technology

Contact Leading Edge Experts & Companies

The Experts below are selected from a list of 360 Experts worldwide ranked by ideXlab platform

Benjamin F Cravatt - One of the best experts on this subject based on the ideXlab platform.

  • inhibiting fatty acid amide hydrolase normalizes endotoxin induced enhanced gastrointestinal motility in mice
    British Journal of Pharmacology, 2012
    Co-Authors: Mohammad Bashashati, Spyros P Nikas, Jodianne T Wood, Shakiru O Alapafuja, Grzegorz Godlewski, Martin Storr, Jie Liu, Catherine M Keenan, Hong Zhang, Benjamin F Cravatt
    Abstract:

    BACKGROUND AND PURPOSE Gastrointestinal (GI) motility is regulated in part by fatty acid ethanolamides (FAEs), including the endocannabinoid (EC) anandamide (AEA). The actions of FAEs are terminated by fatty acid amide hydrolase (FAAH). We investigated the actions of the novel FAAH inhibitor AM3506 on normal and enhanced GI motility. EXPERIMENTAL APPROACH We examined the effect of AM3506 on electrically-evoked contractility in vitro and GI transit and colonic faecal output in vivo, in normal and FAAH-deficient mice treated with saline or LPS (100 µg·kg−1, i.p.), in the presence and absence of cannabinoid (CB) receptor antagonists. mRNA expression was measured by quantitative real time-PCR, EC levels by liquid chromatography-MS and FAAH activity by the conversion of [3H]-AEA to [3H]-ethanolamine in intestinal extracts. FAAH expression was examined by immunohistochemistry. KEY RESULTS FAAH was dominantly expressed in the enteric nervous system; its mRNA levels were higher in the ileum than the colon. LPS enhanced ileal contractility in the absence of overt inflammation. AM3506 reversed the enhanced electrically-evoked contractions of the ileum through CB1 and CB2 receptors. LPS increased the rate of upper GI transit and faecal output. AM3506 normalized the enhanced GI transit through CB1 and CB2 receptors and faecal output through CB1 receptors. LPS did not increase GI transit in FAAH-deficient mice. CONCLUSIONS AND IMPLICATIONS Inhibiting FAAH normalizes various parameters of GI dysmotility in intestinal pathophysiology. Inhibition of FAAH represents a new approach to the treatment of disordered intestinal motility.

  • fatty acid amide hydrolase blockade attenuates the development of collagen induced arthritis and related thermal hyperalgesia in mice
    Pharmacology Biochemistry and Behavior, 2011
    Co-Authors: Benjamin F Cravatt, Steven G Kinsey, Pattipati S Naidu, David T Dudley, Aron H Lichtman
    Abstract:

    Fatty acid amide hydrolase (FAAH) is the primary degradative enzyme of the endocannabinoid anandamide (N-arachidonoylethanolamine), which activates cannabinoid CB(1) and CB(2) receptors. FAAH disruption reduces nociception in a variety of acute rodent models of inflammatory pain. The present study investigated whether these actions extend to the chronic, collagen-induced arthritis (CIA) model. We investigated the anti-arthritic and anti-hyperalgesic effects of genetic deletion or pharmacological inhibition of FAAH in the CIA model. FAAH (-/-) mice, and FAAH-NS mice that express FAAH exclusively in nervous tissue, displayed decreased severity of CIA and associated hyperalgesia. These phenotypic anti-arthritic effects were prevented by repeated daily injections of the CB(2) receptor antagonist, SR144528, but not the CB(1) receptor antagonist rimonabant. Similarly, repeated administration of the FAAH inhibitor URB597 reduced CIA severity, and acute administration of rimonabant, but not SR144528, blocked the anti-hyperalgesic effects of prolonged FAAH inhibition, suggesting that prolonged CB(2) receptor activation reduces the severity of CIA, whereas acute CB(1) receptor activation reduces CIA-induced hyperalgesia. In contrast, acute administration of URB597 elicited a CB(1) receptor-dependent anti-hyperalgesic effect. The observed anti-arthritic and anti-hyperalgesic properties of FAAH inhibition, coupled with a lack of apparent behavioral alterations, suggest that endocannabinoid modulating enzymes offer a promising therapeutic target for the development of novel pharmacological approaches to treat rheumatoid arthritis and associated hyperalgesia.

  • an anatomical and temporal portrait of physiological substrates for fatty acid amide hydrolase
    Journal of Lipid Research, 2011
    Co-Authors: Jonathan Z Long, Melanie Lacava, Benjamin F Cravatt
    Abstract:

    Fatty acid amide hydrolase (FAAH) is an integral membrane serine hydrolase that degrades a number of bioactive lipid amides, including the endocannabinoid anandamide (N-arachidonoyl ethanolamine) (1, 2). Anandamide acts as an endogenous ligand for the CB1 and CB2 receptors (3–6), which are two G-protein-coupled receptors that also respond to Δ9-tetrahydrocannabinol (THC), the psychoactive component of marijuana (7). The genetic or pharmacological inactivation of FAAH results in substantial increases in brain concentrations of anandamide and other N-acyl ethanolamine (NAE) lipids and produces several CB1-dependent neurobehavioral effects in rodents, including anxiolysis (8, 9), anti-depression (10), and anti-nociception (6, 11, 12). Interestingly, these effects are not accompanied by the cognitive and motor dysfunctions associated with direct CB1 agonists such as THC. Taken together, these findings indicate that FAAH is a key regulator of endocannabinoid activity in vivo and suggest further that the enzyme might represent a therapeutic target for the treatment of pain and other nervous system disorders (13, 14). FAAH-disrupted mice have also shown some phenotypes that are not reversed by the administration of CB1 or CB2 antagonists (12, 15, 16), suggesting that anandamide and/or other FAAH substrates possess bioactivities that extend beyond the endocannabinoid system. To more broadly explore the physiological substrate pool regulated by FAAH, our laboratory has analyzed FAAH(−/−) mice using a metabolomics method based on untargeted LC-MS (17). This approach confirmed known elevations in anandamide and other NAEs in brains and livers of FAAH(−/−) mice and also uncovered a novel class of natural products regulated by FAAH, the N-acyl taurines (NATs). Subsequent in vitro studies showed that FAAH can hydrolyze NATs and that NATs are agonists of the transient receptor potential family of ion channels at concentrations approximately equal to or lower than those found in certain tissues from FAAH(−/−) mice (17, 18). Despite the aforementioned advances in our understanding of physiological substrates for FAAH, a comprehensive inventory of these lipids across multiple tissues from wild-type versus FAAH-disrupted animals has not yet been performed. Here, we describe a temporal and anatomical atlas of FAAH substrates following acute (i.e., pharmacological) versus chronic (i.e., genetic) blockade of this enzyme in mice. We find that FAAH control over FA amide substrates is tissue specific and, in some cases, influenced by temporal factors that probably reflect differences in the rate of substrate biosynthesis. These discoveries thus lend further support to the hypothesis that multiple pathways exist for the biosynthesis of both NAEs and NATs in vivo.

  • fatty acid amide hydrolase as a potential therapeutic target for the treatment of pain and cns disorders
    Expert Opinion on Drug Discovery, 2009
    Co-Authors: Douglas S. Johnson, Benjamin F Cravatt
    Abstract:

    Background: Fatty acid amide hydrolase (FAAH) is an integral membrane enzyme that hydrolyzes the endocannabinoid anandamide and related amidated signaling lipids. Genetic or pharmacological inactivation of FAAH produces analgesic, anti-inflammatory, anxiolytic and antidepressant phenotypes without showing the undesirable side effects of direct cannabinoid receptor agonists, indicating that FAAH may be a promising therapeutic target. Objectives: This review highlights advances in the development of FAAH inhibitors of different mechanistic classes and their in vivo efficacy. Also highlighted are advances in technology for the in vitro and in vivo selectivity assessment of FAAH inhibitors using activity-based protein profiling and click chemistry-activity-based protein profiling, respectively. Recent reports on structure-based drug design for human FAAH generated by protein engineering using interspecies active site conversion are also discussed. Methods: The literature searches of Medline and SciFinder data...

  • localization of the endocannabinoid degrading enzyme fatty acid amide hydrolase in rat dorsal root ganglion cells and its regulation after peripheral nerve injury
    The Journal of Neuroscience, 2009
    Co-Authors: Isobel J Lever, Benjamin F Cravatt, Maurice R Elphick, Michelle Robinson, Mario Cibelli, Cleoper C Paule, Peter Santha, Louis Yee, Stephen P Hunt, Istvan Nagy
    Abstract:

    Fatty acid amide hydrolase (FAAH) is a degradative enzyme for a group of endogenous signaling lipids that includes anandamide (AEA). AEA acts as an endocannabinoid and an endovanilloid by activating cannabinoid and vanilloid type 1 transient receptor potential (TRPV1) receptors, respectively, on dorsal root ganglion (DRG) sensory neurons. Inhibition of FAAH activity increases AEA concentrations in nervous tissue and reduces sensory hypersensitivity in animal pain models. Using immunohistochemistry, Western blotting, and reverse transcription-PCR, we demonstrate the location of the FAAH in adult rat DRG, sciatic nerve, and spinal cord. In naive rats, FAAH immunoreactivity localized to the soma of 32.7 ± 0.8% of neurons in L4 and L5 DRG. These were small-sized (mean soma area, 395.96 ± 5.6 μm2) and predominantly colabeled with peripherin and isolectin B4 markers of unmyelinated C-fiber neurons; 68% colabeled with antibodies to TRPV1 (marker of nociceptive DRG neurons), and <2% colabeled with NF200 (marker of large myelinated neurons). FAAH-IR was also present in small, NF200-negative cultured rat DRG neurons. Incubation of these cultures with the FAAH inhibitor URB597 increased AEA-evoked cobalt uptake in a capsazepine-sensitive manner. After sciatic nerve axotomy, there was a rightward shift in the cell-size distribution of FAAH-immunoreactive (IR) DRG neurons ipsilateral to injury: FAAH immunoreactivity was detected in larger-sized cells that colabeled with NF200. An ipsilateral versus contralateral increase in both the size and proportion of FAAH-IR DRG occurred after spinal nerve transection injury but not after chronic inflammation of the rat hindpaw 2 d after injection of complete Freund's adjuvant. This study reveals the location of FAAH in neural tissue involved in peripheral nociceptive transmission.

Hiroko Hama - One of the best experts on this subject based on the ideXlab platform.

  • chapter 60 mouse models of FA2H deficiency
    Movement Disorders (Second Edition)#R##N#Genetics and Models, 2015
    Co-Authors: Kathleen A Willet, Hiroko Hama
    Abstract:

    FA2H encodes a fatty acid 2-hydroxylase essential for the proper functioning of the nervous system. FA2H introduces a 2-hydroxyl group to the N-acyl chain of a major myelin lipid, galactosylceramide. Three types of FA2H knockout mice have been reported to date: two FA2H-null mutants and a conditional mutant lacking FA2H in myelin-forming cells. Although morphologically and functionally normal, myelin is formed in the absence of FA2H, the long-term stability of such myelin is compromised, resulting in gradual demyelination and axonal degeneration. Histological and behavioral studies revealed significant cerebellar abnormalities in FA2H-null mice. The cerebellar deficits are indistinguishable between FA2H-null mice and the conditional mutant lacking FA2H in myelin-forming cells, indicating that myelin lipid abnormalities are the primary cause of these phenotypes. The mouse models will be valuable tools for studies of pathogenesis of FA2H deficiency and in developing therapeutic approaches for this disorder.

  • defective FA2H leads to a novel form of neurodegeneration with brain iron accumulation nbia
    Annals of Neurology, 2010
    Co-Authors: Michael C Kruer, Hiroko Hama, Coro Paisanruiz, Nathalie Boddaert, Moon Y Yoon, Allison Gregory, Alessandro Malandrini, Randall L Woltjer, Arnold Munnich, Stephanie Gobin
    Abstract:

    Objective: Neurodegeneration with brain iron accumulation (NBIA) represents a distinctive phenotype of neurodegenerative disease for which several causative genes have been identified. The spectrum of neurologic disease associated with mutations in NBIA genes is broad, with phenotypes that range from infantile neurodegeneration and death in childhood to adult-onset parkinsonism-dystonia. Here we report the discovery of a novel gene that leads to a distinct form of NBIA. Methods: Using autozygosity mapping and candidate gene sequencing, we identified mutations in the fatty acid hydroxylase gene FA2H, newly implicating abnormalities of ceramide metabolism in the pathogenesis of NBIA. Results: Neuroimaging demonstrated T2 hypointensity in the globus pallidus, confluent T2 white matter hyperintensities, and profound pontocerebellar atrophy in affected members of two families. Phenotypically, affected family members exhibited spastic quadriparesis, ataxia, and dystonia with onset in childhood and episodic neurological decline. Analogous to what has been reported previously for PLA2G6, the phenotypic spectrum of FA2H mutations is diverse based on our findings and those of prior investigators, because FA2H mutations have been identified in both a form of hereditary spastic paraplegia (SPG35) and a progressive familial leukodystrophy. Interpretation: These findings link white matter degeneration and NBIA for the first time and implicate new signaling pathways in the genesis of NBIA. ANN NEUROL 2010;00:000‐000

  • FA2H is responsible for the formation of 2-hydroxy galactolipids in peripheral nervous system myelin
    Journal of lipid research, 2007
    Co-Authors: Eduardo N. Maldonado, Nathan L. Alderson, Paula V. Monje, Patrick M. Wood, Hiroko Hama
    Abstract:

    Myelin in the mammalian nervous system has a high concentration of galactolipids [galactosylceramide (GalCer) and sulfatide] with 2-hydroxy fatty acids. We recently reported that fatty acid 2-hydroxylase (FA2H), encoded by the FA2H gene, is the major fatty acid 2-hydroxylase in the mouse brain. In this report, we show that FA2H also plays a major role in the formation of 2-hydroxy galactolipids in the peripheral nervous system. FA2H mRNA and FA2H activity in the neonatal rat sciatic nerve increased rapidly during developmental myelination. The contents of 2-hydroxy fatty acids were approximately 5% of total galactolipid fatty acids at 4 days of age and increased to 60% in GalCer and to 35% in sulfatides at 60 days of age. The chain length of galactolipid fatty acids also increased significantly during myelination. FA2H expression in cultured rat Schwann cells was highly increased in response to dibutyryl cyclic AMP, which stimulates Schwann cell differentiation and upregulates myelin genes, such as UDP-galactose:ceramide galactosyltransferase and protein zero. These observations indicate that FA2H is a myelination-associated gene. FA2H-directed RNA interference (RNAi) by short-hairpin RNA expression resulted in a reduction of cellular 2-hydroxy fatty acids and 2-hydroxy GalCer in D6P2T Schwannoma cells, providing direct evidence that FA2H-dependent fatty acid 2-hydroxylation is required for the formation of 2-hydroxy galactolipids in peripheral nerve myelin. Interestingly, FA2H-directed RNAi enhanced the migration of D6P2T cells, suggesting that, in addition to their structural role in myelin, 2-hydroxy lipids may greatly influence the migratory properties of Schwann cells.

  • fatty acid 2 hydroxylase encoded by FA2H accounts for differentiation associated increase in 2 oh ceramides during keratinocyte differentiation
    Journal of Biological Chemistry, 2007
    Co-Authors: Yoshikazu Uchida, Nathan L. Alderson, Hiroko Hama, Sounthala Douangpanya, Yu Wang, D Crumrine, Peter M Elias, Walter M Holleran
    Abstract:

    Ceramides in mammalian stratum corneum comprise a heterogeneous mixture of molecular species that subserve the epidermal permeability barrier, an essential function for survival in a terrestrial environment. In addition to a variation of sphingol species, hydroxylation of the amide-linked fatty acids contributes to the diversity of epidermal ceramides. Fatty acid 2-hydroxylase, encoded by the gene FA2H, the mammalian homologue of FAH1 in yeast, catalyzes the synthesis of 2-hydroxy fatty acid-containing sphingolipids. We assessed here whether FA2H accounts for 2-hydroxyceramide/2-hydroxyglucosylceramide synthesis in epidermis. Reverse transcription-PCR and Western immunoblots demonstrated that FA2H is expressed in cultured human keratinocytes and human epidermis, with FA2H expression and fatty acid 2-hydroxylase activity increased with differentiation. FA2H-siRNA suppressed 2-hydroxylase activity and decreased 2-hydroxyceramide/2-hydroxyglucosylceramide levels, demonstrating that FA2H accounts for synthesis of these sphingolipids in keratinocytes. Whereas FA2H expression and 2-hydroxy free fatty acid production increased early in keratinocyte differentiation, production of 2-hydroxyceramides/2-hydroxyglucosylceramides with longer chain amide-linked fatty acids (≥C24) increased later. Keratinocytes transduced with FA2H-siRNA contained abnormal epidermal lamellar bodies and did not form the normal extracellular lamellar membranes required for the epidermal permeability barrier. These results reveal that 1) differentiation-dependent up-regulation of ceramide synthesis and fatty acid elongation is accompanied by up-regulation of FA2H; 2) 2-hydroxylation of fatty acid by FA2H occurs prior to generation of ceramides/glucosylceramides; and 3) 2-hydroxyceramides/2-hydroxyglucosylceramides are required for epidermal lamellar membrane formation. Thus, late differentiation-linked increases in FA2H expression are essential for epidermal permeability barrier homeostasis.

  • FA2H dependent fatty acid 2 hydroxylation in postnatal mouse brain
    Journal of Lipid Research, 2006
    Co-Authors: Nathan L. Alderson, Eduardo N. Maldonado, Michael J. Kern, Narayan R Bhat, Hiroko Hama
    Abstract:

    2-Hydroxy fatty acids are relatively minor species of membrane lipids found almost exclusively as N-acyl chains of sphingolipids. In mammals, 2-hydroxy sphingolipids are uniquely abundant in myelin galactosylceramide and sulfatide. Despite the well-documented abundance of 2-hydroxy galactolipids in the nervous system, the enzymatic process of the 2-hydroxylation is not fully understood. To fill this gap, we have identified a human fatty acid 2-hydroxylase gene (FA2H) that is highly expressed in brain. In this report, we test the hypothesis that FA2H is the major fatty acid 2-hydroxylase in mouse brain and that free 2-hydroxy fatty acids are formed as precursors of myelin 2-hydroxy galactolipids. The fatty acid compositions of galactolipids in neonatal mouse brain gradually changed during the course of myelination. The relative ratio of 2-hydroxy versus nonhydroxy galactolipids was very low at 2 days of age ( approximately 8% of total galactolipids) and increased 6- to 8-fold by 30 days of age. During this period, free 2-hydroxy fatty acid levels in mouse brain increased 5- to 9-fold, and their composition was reflected in the fatty acids in galactolipids, consistent with a precursor-product relationship. The changes in free 2-hydroxy fatty acid levels coincided with fatty acid 2-hydroxylase activity and with the upregulation of FA2H expression. Furthermore, mouse brain fatty acid 2-hydroxylase activity was inhibited by anti-FA2H antibodies. Together, these data provide evidence that FA2H is the major fatty acid 2-hydroxylase in brain and that 2-hydroxylation of free fatty acids is the first step in the synthesis of 2-hydroxy galactolipids.

Pablo Rusjan - One of the best experts on this subject based on the ideXlab platform.

  • lower amygdala fatty acid amide hydrolase in violent offenders with antisocial personality disorder an 11c curb positron emission tomography study
    Translational Psychiatry, 2021
    Co-Authors: Nathan J Kolla, Sylvain Houle, Pablo Rusjan, Isabelle Boileau, Jeremy J Watts, Karolina Karas, Romina Mizrahi
    Abstract:

    Antisocial personality disorder (ASPD) imposes a high societal burden given the repetitive reactive aggression that affected individuals perpetrate. Since the brain endocannabinoid system (ECS) has been implicated in ASPD and aggressive behavior, we utilized [11C]CURB positron emission tomography to investigate fatty acid amide hydrolase (FAAH), an enzyme of the ECS that degrades anandamide, in 16 individuals with ASPD and 16 control participants. We hypothesized that FAAH density would be lower in the amygdala for several reasons. First, decreased FAAH expression is associated with increased cannabinoid receptor 1 stimulation, which may be responsible for amygdala hyper-reactivity in reactive aggression. Second, the amygdala is the seat of the neural circuit mediating reactive aggression. Third, other PET studies of externalizing populations show reduced brain FAAH density. Conversely, we hypothesized that FAAH expression would be greater in the orbitofrontal cortex. Consistent with our hypothesis, we found that amygdala FAAH density was lower in the amygdala of ASPD (p = 0.013). Cerebellar and striatal FAAH expression were inversely related with impulsivity (cerebellum: r = -0.60, p = 0.017; dorsal caudate: r = -0.58, p = 0.023; dorsal putamen: r = -0.55, p = 0.034), while cerebellar FAAH density was also negatively associated with assaultive aggression (r = -0.54, p = 0.035). ASPD presents high levels of disruptive behavior with few, if any, efficacious treatment options. Novel therapeutics that increase FAAH brain levels in a region-specific manner could hold promise for attenuating certain symptom clusters of ASPD, although our results require replication.

  • imaging brain fatty acid amide hydrolase in untreated patients with psychosis
    Biological Psychiatry, 2020
    Co-Authors: Jeremy J Watts, Sylvain Houle, Alan A Wilson, Isabelle Boileau, Rachel F Tyndale, Maya R Jacobson, Nittha Lalang, Michael Kiang, Ruth A Ross, Pablo Rusjan
    Abstract:

    Abstract Background The brain’s endocannabinoid system, the primary target of cannabis, has been implicated in psychosis. The endocannabinoid anandamide is elevated in cerebrospinal fluid of patients with schizophrenia. Fatty acid amide hydrolase (FAAH) controls brain anandamide levels; however, it is unknown if FAAH is altered in vivo in psychosis or related to positive psychotic symptoms. Methods Twenty-seven patients with schizophrenia spectrum disorders and 36 healthy control subjects completed high-resolution positron emission tomography scans with the novel FAAH radioligand [11C]CURB and structural magnetic resonance imaging. Data were analyzed using the validated irreversible 2-tissue compartment model with a metabolite-corrected arterial input function. Results FAAH did not differ significantly between patients with psychotic disorders and healthy control subjects (F1,62.85 = 0.48, p = .49). In contrast, lower FAAH predicted greater positive psychotic symptom severity, with the strongest effect observed for the positive symptom dimension, which includes suspiciousness, delusions, unusual thought content, and hallucinations (F1,26.69 = 12.42, p = .002; Cohen’s f = 0.42, large effect). Shorter duration of illness (F1,26.95 = 13.78, p = .001; Cohen’s f = 0.39, medium to large effect) and duration of untreated psychosis predicted lower FAAH (F1,26.95 = 6.03, p = .021, Cohen’s f = 0.27, medium effect). These results were not explained by past cannabis exposure or current intake of antipsychotic medications. FAAH exhibited marked differences across brain regions (F7,112.62 = 175.85, p 1). Overall, FAAH was higher in female subjects than in male subjects (F1,62.84 = 10.05, p = .002; Cohen’s f = 0.37). Conclusions This first study of brain FAAH in psychosis indicates that FAAH may represent a biomarker of disease state of potential utility for clinical studies targeting psychotic symptoms or as a novel target for interventions to treat psychotic symptoms.

  • fatty acid amide hydrolase binding in brain of cannabis users imaging with the novel radiotracer 11c curb
    Biological Psychiatry, 2016
    Co-Authors: Isabelle Boileau, Pablo Rusjan, Romina Mizrahi, Rachel F Tyndale, Belinda Williams, Esmaeil Mansouri, Bernard Le Foll, Marilyn A Huestis
    Abstract:

    Abstract Background One of the major mechanisms for terminating the actions of the endocannabinoid anandamide is hydrolysis by fatty acid amide hydrolase (FAAH), and inhibitors of the enzyme were suggested as potential treatment for human cannabis dependence. However, the status of brain FAAH in cannabis use disorder is unknown. Methods Brain FAAH binding was measured with positron emission tomography and [ 11 C]CURB in 22 healthy control subjects and ten chronic cannabis users during early abstinence. The FAAH genetic polymorphism (rs324420) and blood, urine, and hair levels of cannabinoids and metabolites were determined. Results In cannabis users, FAAH binding was significantly lower by 14%–20% across the brain regions examined than in matched control subjects (overall Cohen's d = 0.96). Lower binding was negatively correlated with cannabinoid concentrations in blood and urine and was associated with higher trait impulsiveness. Conclusions Lower FAAH binding levels in the brain may be a consequence of chronic and recent cannabis exposure and could contribute to cannabis withdrawal. This effect should be considered in the development of novel treatment strategies for cannabis use disorder that target FAAH and endocannabinoids. Further studies are needed to examine possible changes in FAAH binding during prolonged cannabis abstinence and whether lower FAAH binding predates drug use.

  • the fatty acid amide hydrolase c385a variant affects brain binding of the positron emission tomography tracer 11c curb
    Journal of Cerebral Blood Flow and Metabolism, 2015
    Co-Authors: Isabelle Boileau, Pablo Rusjan, Rachel F Tyndale, Belinda Williams, Esmaeil Mansouri, Duncan Westwood, Bernard Le Foll, Romina Mizrahi
    Abstract:

    The common functional single-nucleotide polymorphism (rs324420, C385A) of the endocannabinoid inactivating enzyme fatty acid amide hydrolase (FAAH) has been associated with anxiety disorder relevant phenotype and risk for addictions. Here, we tested whether the FAAH polymorphism affects in vivo binding of the FAAH positron emission tomography (PET) probe [11C]CURB ([11C-carbonyl]-6-hydroxy-[1,10-biphenyl]-3-yl cyclohexylcarbamate (URB694)). Participants (n=24) completed one [11C]CURB/PET scan and were genotyped for rs324420. Relative to C/C (58%), A-allele carriers (42%) had 23% lower [11C]CURB binding (λk3) in brain. We report evidence that the genetic variant rs324420 in FAAH is associated with measurable differences in brain FAAH binding as per PET [11C]CURB measurement.

Matthew P Patricelli - One of the best experts on this subject based on the ideXlab platform.

  • comparative characterization of a wild type and transmembrane domain deleted fatty acid amide hydrolase identification of the transmembrane domain as a site for oligomerization
    Biochemistry, 1998
    Co-Authors: Matthew P Patricelli, Hilal A Lashuel, Dan K Giang, Jeffery W Kelly, Benjamin F Cravatt
    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 (ΔTM-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 ΔTM-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 ΔTM-FAAH by chemical cross-linking, sedimentation velocity analytical ultracentrifugation, and size exclusion chromatography indicated that both enzymes were oligomeric when membra...

  • comparative characterization of a wild type and transmembrane domain deleted fatty acid amide hydrolase identification of the transmembrane domain as a site for oligomerization
    Biochemistry, 1998
    Co-Authors: Matthew P Patricelli, Hilal A Lashuel, Dan K Giang, Jeffery W Kelly, Benjamin F Cravatt
    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.

  • an endogenous sleep inducing compound is a novel competitive inhibitor of fatty acid amide hydrolase
    Bioorganic & Medicinal Chemistry Letters, 1998
    Co-Authors: Matthew P Patricelli, Dale L Boger, Jean E. Patterson, Benjamin F Cravatt
    Abstract:

    Abstract 2-Octyl γ-bromoacetoacetate (OγBr), 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 OγBr and its reported sleep-inducing effects, we synthesized OγBr 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. OγBr was found to competitively inhibit FAAH activity with IC 50 and K i values of 2.6 μM and 0.8 μM, respectively [for the ( R )-enantiomer of OγBr ( 1 )]. A set of synthetic analogs of OγBr 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 γ-halo β-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 OγBr may be a physiological regulator of FAAH activity and fatty acid amide levels in vivo. Additionally, the characterization of γ-halo β-keto esters as powerful FAAH inhibitors near physiological pH may aid in future studies of the enzymology and biological properties of FAAH.

Dan K Giang - One of the best experts on this subject based on the ideXlab platform.

  • comparative characterization of a wild type and transmembrane domain deleted fatty acid amide hydrolase identification of the transmembrane domain as a site for oligomerization
    Biochemistry, 1998
    Co-Authors: Matthew P Patricelli, Hilal A Lashuel, Dan K Giang, Jeffery W Kelly, Benjamin F Cravatt
    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 (ΔTM-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 ΔTM-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 ΔTM-FAAH by chemical cross-linking, sedimentation velocity analytical ultracentrifugation, and size exclusion chromatography indicated that both enzymes were oligomeric when membra...

  • comparative characterization of a wild type and transmembrane domain deleted fatty acid amide hydrolase identification of the transmembrane domain as a site for oligomerization
    Biochemistry, 1998
    Co-Authors: Matthew P Patricelli, Hilal A Lashuel, Dan K Giang, Jeffery W Kelly, Benjamin F Cravatt
    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.

  • molecular characterization of human and mouse fatty acid amide hydrolases
    Proceedings of the National Academy of Sciences of the United States of America, 1997
    Co-Authors: Dan K Giang, Benjamin F Cravatt
    Abstract:

    Recently, we reported the isolation, cloning, and expression of a rat enzyme, fatty acid amide hydrolase (FAAH), that degrades bioactive fatty acid amides like oleamide and anandamide to their corresponding acids, thereby serving to terminate the signaling functions of these molecules. Here, we report the molecular characterization of both a mouse and a human FAAH and compare these enzymes to the rat FAAH. The enzymes are well conserved in primary structure, with the mouse and rat FAAHs sharing 91% amino acid identity and the human FAAH sharing 82% and 84% identity with the rat FAAH and mouse FAAH, respectively. In addition, the expressed human and rat FAAHs behave biochemically as membrane proteins of comparable molecular size and show similar, but distinguishable, enzymological properties. The identification of highly homologous FAAH proteins in rat, mouse, and human supports a general role for the fatty acid amides in mammalian biology.