Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
PLoS One ; 17(9): e0274774, 2022.
Article in English | MEDLINE | ID: mdl-36126056

ABSTRACT

Methylmalonic acidemia (MMA) is an inborn error of metabolism mostly caused by mutations in the mitochondrial methylmalonyl-CoA mutase gene (MMUT). MMA patients suffer from frequent episodes of metabolic decompensation, which can be life threatening. To mimic both the dietary restrictions and metabolic decompensation seen in MMA patients, we developed a novel protein-controlled diet regimen in a Mmut deficient mouse model of MMA and demonstrated the therapeutic benefit of mLB-001, a nuclease-free, promoterless recombinant AAV GeneRideTM vector designed to insert the mouse Mmut into the endogenous albumin locus via homologous recombination. A single intravenous administration of mLB-001 to neonatal or adult MMA mice prevented body weight loss and mortality when challenged with a high protein diet. The edited hepatocytes expressed functional MMUT protein and expanded over time in the Mmut deficient mice, suggesting a selective growth advantage over the diseased cells. In mice with a humanized liver, treatment with a human homolog of mLB-001 resulted in site-specific genome editing and transgene expression in the transplanted human hepatocytes. Taken together, these findings support the development of hLB-001 that is currently in clinical trials in pediatric patients with severe forms of MMA.


Subject(s)
Amino Acid Metabolism, Inborn Errors , Methylmalonyl-CoA Mutase , Adult , Albumins/genetics , Amino Acid Metabolism, Inborn Errors/genetics , Amino Acid Metabolism, Inborn Errors/therapy , Animals , Child , Disease Models, Animal , Gene Editing , Humans , Methylmalonyl-CoA Mutase/genetics , Methylmalonyl-CoA Mutase/metabolism , Mice
2.
Cell Mol Immunol ; 18(6): 1463-1475, 2021 06.
Article in English | MEDLINE | ID: mdl-31797905

ABSTRACT

His-tRNA synthetase (HARS) is targeted by autoantibodies in chronic and acute inflammatory anti-Jo-1-positive antisynthetase syndrome. The extensive activation and migration of immune cells into lung and muscle are associated with interstitial lung disease, myositis, and morbidity. It is unknown whether the sequestration of HARS is an epiphenomenon or plays a causal role in the disease. Here, we show that HARS circulates in healthy individuals, but it is largely undetectable in the serum of anti-Jo-1-positive antisynthetase syndrome patients. In cultured primary human skeletal muscle myoblasts (HSkMC), HARS is released in increasing amounts during their differentiation into myotubes. We further show that HARS regulates immune cell engagement and inhibits CD4+ and CD8+ T-cell activation. In mouse and rodent models of acute inflammatory diseases, HARS administration downregulates immune activation. In contrast, neutralization of extracellular HARS by high-titer antibody responses during tissue injury increases susceptibility to immune attack, similar to what is seen in humans with anti-Jo-1-positive disease. Collectively, these data suggest that extracellular HARS is homeostatic in normal subjects, and its sequestration contributes to the morbidity of the anti-Jo-1-positive antisynthetase syndrome.


Subject(s)
Histidine-tRNA Ligase/blood , Immunity , Organ Specificity , Animals , Autoantibodies/blood , Case-Control Studies , Cell Differentiation/drug effects , Disease Models, Animal , Female , Histidine-tRNA Ligase/immunology , Humans , Immunity/drug effects , Immunomodulation/drug effects , Insulin-Like Growth Factor I/pharmacology , Lung/drug effects , Lung/pathology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Male , Mice, Inbred C57BL , Middle Aged , Muscle Cells/drug effects , Muscle Cells/enzymology , Muscles/drug effects , Muscles/pathology , Myositis/blood , Myositis/diagnostic imaging , Myositis/immunology , Organ Specificity/drug effects , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Tomography, X-Ray Computed
3.
Hepatology ; 73(6): 2223-2237, 2021 06.
Article in English | MEDLINE | ID: mdl-32976669

ABSTRACT

BACKGROUND AND AIMS: Adeno-associated viral (AAV) gene therapy has shown great promise as an alternative treatment for metabolic disorders managed using liver transplantation, but remains limited by transgene loss and genotoxicity. Our study aims to test an AAV vector with a promoterless integrating cassette, designed to provide sustained hepatic transgene expression and reduced toxicity in comparison to canonical AAV therapy. APPROACH AND RESULTS: Our AAV vector was designed to insert a methylmalonyl-CoA mutase (MMUT) transgene into the 3' end of the albumin locus and tested in mouse models of methylmalonic acidemia (MMA). After neonatal delivery, we longitudinally evaluated hepatic transgene expression, plasma levels of methylmalonate, and the MMA biomarker, fibroblast growth factor 21 (Fgf21), as well as integration of MMUT in the albumin locus. At necropsy, we surveyed for AAV-related hepatocellular carcinoma (HCC) in all treated MMA mice and control littermates. AAV-mediated genome editing of MMUT into the albumin locus resulted in permanent hepatic correction in MMA mouse models, which was accompanied by decreased levels of methylmalonate and Fgf21, and improved survival without HCC. With time, levels of transgene expression increased and methylmalonate progressively decreased, whereas the number of albumin-MMUT integrations and corrected hepatocytes in MMA mice increased, but not in similarly treated wild-type animals. Additionally, expression of MMUT in the setting of MMA conferred a selective growth advantage upon edited cells, which potentiates the therapeutic response. CONCLUSIONS: In conclusion, our findings demonstrate that AAV-mediated, promoterless, nuclease-free genome editing at the albumin locus provides safe and durable therapeutic benefit in neonatally treated MMA mice.


Subject(s)
Amino Acid Metabolism, Inborn Errors/therapy , Dependovirus/genetics , Gene Editing/methods , Genetic Therapy/methods , Methylmalonyl-CoA Mutase/metabolism , Amino Acid Metabolism, Inborn Errors/metabolism , Animals , Animals, Newborn , Biomarkers/blood , Carcinoma, Hepatocellular/pathology , Disease Models, Animal , Fibroblast Growth Factors/blood , Hepatocytes , Liver Neoplasms/pathology , Liver Transplantation , Malonates/blood , Methylmalonyl-CoA Mutase/genetics , Mice , Mice, Inbred C57BL
4.
Science ; 345(6194): 328-32, 2014 Jul 18.
Article in English | MEDLINE | ID: mdl-25035493

ABSTRACT

Genetic efficiency in higher organisms depends on mechanisms to create multiple functions from single genes. To investigate this question for an enzyme family, we chose aminoacyl tRNA synthetases (AARSs). They are exceptional in their progressive and accretive proliferation of noncatalytic domains as the Tree of Life is ascended. Here we report discovery of a large number of natural catalytic nulls (CNs) for each human AARS. Splicing events retain noncatalytic domains while ablating the catalytic domain to create CNs with diverse functions. Each synthetase is converted into several new signaling proteins with biological activities "orthogonal" to that of the catalytic parent. We suggest that splice variants with nonenzymatic functions may be more general, as evidenced by recent findings of other catalytically inactive splice-variant enzymes.


Subject(s)
Amino Acyl-tRNA Synthetases/metabolism , Catalytic Domain , Alternative Splicing , Amino Acyl-tRNA Synthetases/chemistry , Amino Acyl-tRNA Synthetases/genetics , Catalysis , Humans , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Organ Specificity , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
5.
J Biol Chem ; 289(28): 19269-75, 2014 Jul 11.
Article in English | MEDLINE | ID: mdl-24898250

ABSTRACT

Inflammatory and debilitating myositis and interstitial lung disease are commonly associated with autoantibodies (anti-Jo-1 antibodies) to cytoplasmic histidyl-tRNA synthetase (HisRS). Anti-Jo-1 antibodies from different disease-afflicted patients react mostly with spatially separated epitopes in the three-dimensional structure of human HisRS. We noted that two HisRS splice variants (SVs) include these spatially separated regions, but each SV lacks the HisRS catalytic domain. Despite the large deletions, the two SVs cross-react with a substantial population of anti-Jo-l antibodies from myositis patients. Moreover, expression of at least one of the SVs is up-regulated in dermatomyositis patients, and cell-based experiments show that both SVs and HisRS can be secreted. We suggest that, in patients with inflammatory myositis, anti-Jo-1 antibodies may have extracellular activity.


Subject(s)
Alternative Splicing , Autoantibodies/metabolism , Epitopes/metabolism , Histidine-tRNA Ligase/metabolism , Myositis/enzymology , Autoantibodies/immunology , Cell Line, Tumor , Epitopes/genetics , Epitopes/immunology , Histidine-tRNA Ligase/genetics , Histidine-tRNA Ligase/immunology , Humans , Inflammation/enzymology , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Myositis/genetics , Myositis/immunology , Myositis/pathology , Protein Structure, Tertiary
6.
Structure ; 20(9): 1470-7, 2012 Sep 05.
Article in English | MEDLINE | ID: mdl-22958643

ABSTRACT

Aminoacyl-tRNA synthetases (AARSs) catalyze aminoacylation of tRNAs in the cytoplasm. Surprisingly, AARSs also have critical extracellular and nuclear functions. Evolutionary pressure for new functions might be manifested by splice variants that skip only an internal catalytic domain (CD) and link noncatalytic N- and C-terminal polypeptides. Using disease-associated histidyl-tRNA synthetase (HisRS) as an example, we found an expressed 171-amino acid protein (HisRSΔCD) that deleted the entire CD, and joined an N-terminal WHEP to the C-terminal anticodon-binding domain (ABD). X-ray crystallography and three-dimensional NMR revealed the structures of human HisRS and HisRSΔCD. In contrast to homodimeric HisRS, HisRSΔCD is monomeric, where rupture of the ABD's packing with CD resulted in a dumbbell-like structure of flexibly linked WHEP and ABD domains. In addition, the ABD of HisRSΔCD presents a distinct local conformation. This natural internally deleted HisRS suggests evolutionary pressure to reshape AARS tertiary and quaternary structures for repurposing.


Subject(s)
Evolution, Molecular , Histidine-tRNA Ligase/chemistry , Sequence Deletion , Antibodies/blood , Antibodies/immunology , Base Sequence , Catalytic Domain , Crystallography, X-Ray , DNA, Complementary/genetics , High-Throughput Nucleotide Sequencing , Histidine-tRNA Ligase/genetics , Histidine-tRNA Ligase/immunology , Humans , Lung Diseases, Interstitial/blood , Lung Diseases, Interstitial/immunology , Models, Molecular , Molecular Sequence Data , Myositis/blood , Myositis/immunology , Protein Isoforms , Protein Structure, Secondary , Sequence Analysis, DNA , Transcriptome
7.
J Lipid Res ; 51(10): 2896-908, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20625037

ABSTRACT

Cholesteryl ester (CE) accumulation in macrophages represents a crucial event during foam cell formation, a hallmark of atherogenesis. Here we investigated the role of two previously described CE hydrolases, hormone-sensitive lipase (HSL) and KIAA1363, in macrophage CE hydrolysis. HSL and KIAA1363 exhibited marked differences in their abilities to hydrolyze CE, triacylglycerol (TG), diacylglycerol (DG), and 2-acetyl monoalkylglycerol ether (AcMAGE), a precursor for biosynthesis of platelet-activating factor (PAF). HSL efficiently cleaved all four substrates, whereas KIAA1363 hydrolyzed only AcMAGE. This contradicts previous studies suggesting that KIAA1363 is a neutral CE hydrolase. Macrophages of KIAA1363(-/-) and wild-type mice exhibited identical neutral CE hydrolase activity, which was almost abolished in tissues and macrophages of HSL(-/-) mice. Conversely, AcMAGE hydrolase activity was diminished in macrophages and some tissues of KIAA1363(-/-) but unchanged in HSL(-/-) mice. CE turnover was unaffected in macrophages lacking KIAA1363 and HSL, whereas cAMP-dependent cholesterol efflux was influenced by HSL but not by KIAA1363. Despite decreased CE hydrolase activities, HSL(-/-) macrophages exhibited CE accumulation similar to wild-type (WT) macrophages. We conclude that additional enzymes must exist that cooperate with HSL to regulate CE levels in macrophages. KIAA1363 affects AcMAGE hydrolase activity but is of minor importance as a direct CE hydrolase in macrophages.


Subject(s)
Cholesterol Esters/metabolism , Macrophages/enzymology , Serine Proteases/genetics , Sterol Esterase/genetics , Animals , Atherosclerosis/genetics , Atherosclerosis/metabolism , COS Cells , Chlorocebus aethiops , Diglycerides/metabolism , Hydrolysis , Macrophages/metabolism , Mice , Mice, Inbred Strains , Microscopy, Fluorescence , Serine Proteases/metabolism , Sterol Esterase/metabolism , Transfection
8.
ACS Chem Biol ; 4(11): 958-68, 2009 Nov 20.
Article in English | MEDLINE | ID: mdl-19799466

ABSTRACT

Post-translational modification of histones plays an integral role in regulation of genomic expression through modulation of chromatin structure and function. Chemical preparations of histones bearing these modifications allows for comprehensive in vitro mechanistic investigation into their action to deconvolute observations from genome-wide studies in vivo. Previously, we reported the semisynthesis of ubiquitylated histone H2B (uH2B) using two orthogonal expressed protein ligation reactions. Semisynthetic uH2B, when incorporated into nucleosomes, directly stimulates methylation of histone H3 lysine 79 (K79) by the methyltransferase, disruptor of telomeric silencing-like (Dot1L). Although recruitment of Dot1L to the nucleosomal surface by uH2B could be excluded, comprehensive mechanistic analysis was precluded by systematic limitations in the ability to generate uH2B in large scale. Here we report a highly optimized synthesis of ubiquitylated H2B bearing a G76A point mutation u(G76A)H2B, yielding tens of milligrams of ubiquitylated protein. u(G76A)H2B is indistinguishable from the native uH2B by Dot1L, allowing for detailed studies of the resultant trans-histone crosstalk. Kinetic and structure-activity relationship analyses using u(G76A)H2B suggest a noncanonical role for ubiquitin in the enhancement of the chemical step of H3K79 methylation. Furthermore, titration of the level of uH2B within the nucleosome revealed a 1:1 stoichiometry of Dot1L activation.


Subject(s)
Histones/metabolism , Methyltransferases/metabolism , Nucleosomes/metabolism , Ubiquitinated Proteins/metabolism , Xenopus/metabolism , Animals , Histone-Lysine N-Methyltransferase , Histones/chemistry , Histones/genetics , Humans , Kinetics , Methylation , Methyltransferases/genetics , Models, Molecular , Mutation , Protein Structure, Quaternary , Protein Structure, Tertiary
9.
Chem Biol ; 16(9): 1001-12, 2009 Sep 25.
Article in English | MEDLINE | ID: mdl-19778728

ABSTRACT

Cells control their own death through a program termed apoptosis, which is indispensable for development and homeostasis in all metazoans. Lysosomal cysteine proteases are not normally thought of as participating in apoptosis; however, recent reports have shown that the cathepsin proteases can be released from the lysosome during apoptosis, where they can participate in cell death. We report here the development of an activity-based probe that, under optimized conditions, reports on cathepsin B activity only in apoptotic cells by reading out the release of cathepsin B from the lysosomes. Biochemical characterization of apoptosis in cells from cathepsin B null mice shows delayed and suboptimal activation of caspases. Our data further supports a role for cathepsin B in the cytosol as a positive regulator of a cell death feed-forward loop and provides a chemical tool for future investigations.


Subject(s)
Apoptosis , Carbamates/pharmacology , Cathepsin B/metabolism , Oligopeptides/pharmacology , Amino Acid Sequence , Animals , Biotin/chemistry , Biotin/metabolism , Carbamates/chemistry , Caspases/chemistry , Caspases/metabolism , Cathepsin B/analysis , Cell Line, Tumor , Cytosol/metabolism , Humans , Lysosomes/metabolism , Mice , Molecular Sequence Data , NIH 3T3 Cells , Oligopeptides/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
11.
Sci Signal ; 1(42): pe45, 2008 Oct 21.
Article in English | MEDLINE | ID: mdl-18941136

ABSTRACT

Complete understanding of the molecular mechanisms of signal transduction is a major focus of research in the postgenomic era. Efforts to realize this goal have benefited from interdisciplinary approaches, specifically through the development of new technologies. Several groups have now described new chemical strategies to probe signaling with broadly different degrees of focus. The first report employs an enzymatic tagging strategy to monitor global proteolytic cleavage events that occur during apoptosis. The second study illustrates a general method to functionally characterize a specific kinase through the identification of its substrates. Finally, the third report describes a synthetic route to generate ubiquitinated proteins and demonstrates the utility of protein chemistry in understanding the detailed biochemical mechanisms of signaling. Together, these papers highlight the power of chemical tools to examine specific networks, thereby expanding our understanding of signaling pathways.


Subject(s)
Apoptosis/physiology , Protein Kinases/metabolism , Signal Transduction/physiology , Ubiquitination/physiology , Animals , Biomedical Research/methods , Biomedical Research/trends , Humans , Substrate Specificity/physiology
12.
Chem Biol ; 13(10): 1041-50, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17052608

ABSTRACT

Hundreds, if not thousands, of uncharacterized enzymes currently populate the human proteome. Assembly of these proteins into the metabolic and signaling pathways that govern cell physiology and pathology constitutes a grand experimental challenge. Here, we address this problem by using a multidimensional profiling strategy that combines activity-based proteomics and metabolomics. This approach determined that KIAA1363, an uncharacterized enzyme highly elevated in aggressive cancer cells, serves as a central node in an ether lipid signaling network that bridges platelet-activating factor and lysophosphatidic acid. Biochemical studies confirmed that KIAA1363 regulates this pathway by hydrolyzing the metabolic intermediate 2-acetyl monoalkylglycerol. Inactivation of KIAA1363 disrupted ether lipid metabolism in cancer cells and impaired cell migration and tumor growth in vivo. The integrated molecular profiling method described herein should facilitate the functional annotation of metabolic enzymes in any living system.


Subject(s)
Carboxylic Ester Hydrolases/metabolism , Ovarian Neoplasms/metabolism , Phospholipid Ethers/metabolism , Carbamates/chemistry , Carbamates/pharmacology , Carboxylic Ester Hydrolases/antagonists & inhibitors , Carboxylic Ester Hydrolases/chemistry , Cell Line, Tumor , Chromatography, High Pressure Liquid/methods , Enzyme Activation/drug effects , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Female , Humans , Ketones/chemistry , Ketones/pharmacology , Lysophospholipids/chemistry , Lysophospholipids/metabolism , Ovarian Neoplasms/enzymology , Phospholipid Ethers/chemistry , Platelet Activating Factor/metabolism , RNA Interference , Sensitivity and Specificity , Signal Transduction/drug effects , Sterol Esterase
13.
Chem Res Toxicol ; 19(9): 1142-50, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16978018

ABSTRACT

Serine hydrolase KIAA1363 is highly expressed in invasive cancer cells and is the major protein in mouse brain diethylphosphorylated by and hydrolyzing low levels of chlorpyrifos oxon (CPO) (the activated metabolite of a major insecticide). It is also the primary CPO-hydrolyzing enzyme in spinal cord, kidney, heart, lung, testis, and muscle but not liver, a pattern of tissue expression confirmed by fluorophosphonate-rhodamine labeling. KIAA1363 gene deletion using homologous recombination reduces CPO binding, hydrolysis, and metabolism 3-29-fold on incubation with brain membranes and homogenates determined with 1 nM [(3)H-ethyl]CPO and the inhibitory potency for residual CPO with butyrylcholinesterase as a biomarker. Studies with knockout mice further show that KIAA1363 partially protects brain AChE and monoacylglycerol lipase from CPO-induced in vivo inhibition. Surprisingly, mouse brain KIAA1363 and AChE are similar in in vitro sensitivity to seven methyl, ethyl, and propyl but not higher alkyl OP insecticides and analogues, prompting structural comparisons of the active sites of KIAA1363 and AChE relative to OP potency and selectivity. Homology modeling based largely on the Archaeoglobus fulgidus esterase crystal structure indicates that KIAA1363 has a catalytic triad of S191, D348, and H378, a GDSAG motif, and an oxyanion hole of H113, G114, G115, and G116. Excellent selectivity for KIAA1363 is achieved on OP structure optimization with long alkyl chain substituents suggesting that KIAA1363 has larger acyl and leaving group pockets than those of AChE. KIAA1363 reactivates faster than AChE presumably due to differences in the uncoupling of the catalytic triad His upon phosphorylation. The structural modeling of KIAA1363 helps us understand OP structure-activity relationships and the toxicological relevance of this detoxifying enzyme.


Subject(s)
Chlorpyrifos/analogs & derivatives , Organophosphorus Compounds/toxicity , Serine Endopeptidases/metabolism , Animals , Binding Sites , Chlorpyrifos/metabolism , Chlorpyrifos/toxicity , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/toxicity , Female , Male , Mice , Mice, Inbred C57BL , Models, Molecular , Organophosphorus Compounds/metabolism , Serine Endopeptidases/chemistry , Serine Endopeptidases/genetics , Serine Proteases , Serine Proteinase Inhibitors , Sterol Esterase
14.
J Am Chem Soc ; 127(18): 6662-71, 2005 May 11.
Article in English | MEDLINE | ID: mdl-15869287

ABSTRACT

Transthyretin (TTR) amyloidogenesis requires rate-limiting tetramer dissociation and partial monomer denaturation to produce a misassembly competent species. This process has been followed by turbidity to identify transthyretin amyloidogenesis inhibitors including dibenzofuran-4,6-dicarboxylic acid (1). An X-ray cocrystal structure of TTR.1(2) reveals that it only utilizes the outer portion of the two thyroxine binding pockets to bind to and inhibit TTR amyloidogenesis. Herein, structure-based design was employed to append aryl substituents at C1 of the dibenzofuran ring to complement the unused inner portion of the thyroxine binding pockets. Twenty-eight amyloidogenesis inhibitors of increased potency and dramatically increased plasma TTR binding selectivity resulted. These function by imposing kinetic stabilization on the native tetrameric structure of TTR, creating a barrier that is insurmountable under physiological conditions. Since kinetic stabilization of the TTR native state by interallelic trans suppression is known to ameliorate disease, there is reason to be optimistic that the dibenzofuran-based inhibitors will do the same. Preventing the onset of amyloidogenesis is the most conservative strategy to intervene clinically, as it remains unclear which of the TTR misassembly intermediates results in toxicity. The exceptional binding selectivity enables these inhibitors to occupy the thyroxine binding site(s) in a complex biological fluid such as blood plasma, required for inhibition of amyloidogenesis in humans. It is now established that the dibenzofuran-based amyloidogenesis inhibitors have high selectivity, affinity, and efficacy and are thus excellent candidates for further pharmacologic evaluation.


Subject(s)
Amyloid/antagonists & inhibitors , Benzofurans/chemistry , Benzofurans/pharmacology , Prealbumin/antagonists & inhibitors , Amyloid/biosynthesis , Amyloid/chemistry , Dicarboxylic Acids/chemistry , Dicarboxylic Acids/pharmacology , Kinetics , Models, Molecular , Prealbumin/chemistry , Prealbumin/metabolism , Structure-Activity Relationship , Substrate Specificity
15.
Proc Natl Acad Sci U S A ; 102(17): 6195-200, 2005 Apr 26.
Article in English | MEDLINE | ID: mdl-15840715

ABSTRACT

Organophosphorus (OP) insecticides and chemical warfare agents act primarily by inhibiting acetylcholinesterase. There are many secondary targets for OP toxicants as observed for example with the major insecticide chlorpyrifos and its bioactivated metabolite chlorpyrifos oxon (CPO). Therefore, it was surprising that the predominant mouse brain protein labeled in vitro by [(3)H-ethyl]CPO (1 nM) (designated CPO-binding protein or CPO-BP) is not one of these known OP toxicant targets. CPO-BP is a 50-kDa membrane-bound serine hydrolase measured by derivatization with [(3)H]CPO and SDS/PAGE or filtration binding assay. It appears to undergo rapid diethylphosphorylation by [(3)H]CPO followed by either dephosphorylation and reactivation or aging on loss of an ethyl group. CPO and several other OP toxicants potently inhibit CPO-BP in vivo (i.p., 2 h) (50% inhibition at 2-25 mg/kg) and in vitro (50% inhibition at 8-68 nM). Using three chemical labeling reagents, i.e., [(3)H]CPO and the activity-based proteomic probes fluorophosphonate-biotin and fluorophosphonate-rhodamine, mouse brain CPO-BP is identified as serine hydrolase KIAA1363 of unknown function. Brains from KIAA1363(-/-) mice show greatly reduced levels of CPO labeling and hydrolytic metabolism compared to brains from wild-type mice. KIAA1363 therefore is the principal enzyme for metabolizing low levels of CPO in brain and may play a more general role in detoxification of OP nerve poisons.


Subject(s)
Brain/enzymology , Chlorpyrifos/analogs & derivatives , Chlorpyrifos/pharmacokinetics , Chlorpyrifos/toxicity , Neurotoxins/pharmacokinetics , Animals , Cell Membrane/metabolism , Hydrolysis , Inactivation, Metabolic , Kinetics , Male , Mice , Serine Endopeptidases , Serine Proteases , Sterol Esterase , Tritium
16.
J Med Chem ; 48(5): 1576-87, 2005 Mar 10.
Article in English | MEDLINE | ID: mdl-15743199

ABSTRACT

Amyloid fibril formation by the plasma protein transthyretin (TTR), requiring rate-limiting tetramer dissociation and monomer misfolding, is implicated in several human diseases. Amyloidogenesis can be inhibited through native state stabilization, mediated by small molecule binding to TTR's primarily unoccupied thyroid hormone binding sites. New native state stabilizers have been discovered herein by the facile condensation of arylaldehydes with aryloxyamines affording a bisarylaldoxime ether library. Of the library's 95 compounds, 31 were active inhibitors of TTR amyloid formation in vitro. The bisaryloxime ethers selectively stabilize the native tetrameric state of TTR over the dissociative transition state under amyloidogenic conditions, leading to an increase in the dissociation activation barrier. Several bisaryloxime ethers bind selectively to TTR in human blood plasma over the plethora of other plasma proteins, a necessary attribute for efficacy in vivo. While bisarylaldoxime ethers are susceptible to degradation by N-O bond cleavage, this process is slowed by their binding to TTR. Furthermore, the degradation rate of many of the bisarylaldoxime ethers is slow relative to the half-life of plasma TTR. The bisaryloxime ether library provides valuable structure-activity relationship insight for the development of structurally analogous inhibitors with superior stability profiles, should that prove necessary.


Subject(s)
Amyloid/antagonists & inhibitors , Ethers/chemical synthesis , Hydrazines/chemical synthesis , Oximes/chemical synthesis , Prealbumin/antagonists & inhibitors , Amyloid/metabolism , Crystallography, X-Ray , Drug Stability , Ethers/blood , Ethers/chemistry , Humans , Hydrazines/blood , Hydrazines/chemistry , In Vitro Techniques , Oximes/blood , Oximes/chemistry , Prealbumin/metabolism , Protein Binding , Protein Structure, Quaternary , Structure-Activity Relationship , Ultracentrifugation
17.
Bioorg Med Chem Lett ; 15(4): 1075-8, 2005 Feb 15.
Article in English | MEDLINE | ID: mdl-15686915

ABSTRACT

Ten oxazoles bearing a C(4) carboxyl group were synthesized and evaluated as transthyretin (TTR) amyloid fibril inhibitors. Substituting aryls at the C(2) position of the oxazole ring reveals that a 3,5-dichlorophenyl substituent significantly reduced amyloidogenesis. The efficacy of these inhibitors was enhanced further by installing an ethyl, a propyl, or a CF(3) group at the C(5) position. The CF(3) substitution at C(5) also improves the TTR binding selectivity over all the other proteins in human blood.


Subject(s)
Amyloid/antagonists & inhibitors , Oxazoles/chemical synthesis , Prealbumin/chemical synthesis , Amyloid/biosynthesis , Blood Proteins/metabolism , Drug Design , Humans , Oxazoles/pharmacology , Prealbumin/pharmacology , Structure-Activity Relationship
18.
Hum Mol Genet ; 13(18): 2113-9, 2004 Sep 15.
Article in English | MEDLINE | ID: mdl-15254019

ABSTRACT

Fatty acid amide hydrolase (FAAH) inactivates the endogenous cannabinoid (endocannabinoid) anandamide and related lipid transmitters in vivo. A single nucleotide polymorphism (SNP) in the human FAAH gene (385C to A) has recently been described that, in homozygous form, is over-represented in subjects with problem drug use. This SNP, which converts a conserved proline residue in FAAH to threonine (P129T), suggests a potential role for the FAAH-endocannabinoid system in regulating addictive behavior. Nonetheless, the impact of the 385A mutation on the biochemical and cellular function of FAAH remains unknown. Here, we report that T-lymphocytes isolated from patients homozygous for the P129T-FAAH variant express less than half of the FAAH protein and activity observed in wild-type (WT) lymphocytes. Transfected COS-7 cells also expressed significantly lower levels of P129T-FAAH compared with WT-FAAH, indicating that the aberrant expression of the former protein is not a cell type-specific phenomenon. A comparison of the transcription/translation efficiencies and cellular stabilities of WT- and P129T-FAAH proteins revealed that the reduced expression of the mutant enzyme is due to a post-translational mechanism that precedes productive folding. These findings indicate that the natural 385A SNP in the human FAAH gene produces a mutant enzyme with reduced cellular stability, thus fortifying a potential link between functional abnormalities in the endocannabinoid system and drug abuse and dependence.


Subject(s)
Amidohydrolases/genetics , Amidohydrolases/metabolism , Substance-Related Disorders/enzymology , Substance-Related Disorders/genetics , Amidohydrolases/analysis , Amino Acid Sequence , Animals , COS Cells , Cannabinoid Receptor Modulators/metabolism , Chlorocebus aethiops , Endocannabinoids , Gene Expression , Half-Life , Humans , Molecular Sequence Data , Mutation/genetics , Polymorphism, Single Nucleotide , Proteasome Endopeptidase Complex/metabolism , Protein Conformation , T-Lymphocytes/chemistry , T-Lymphocytes/metabolism , Transfection
20.
Proc Natl Acad Sci U S A ; 99(12): 8394-9, 2002 Jun 11.
Article in English | MEDLINE | ID: mdl-12060782

ABSTRACT

Problem drug use and dependence are neurobehavioral disorders of complex origin. Although environmental factors contribute to drug abuse and addiction, genetic factors also play a significant role estimated at 40-60% of the total risk. Nonetheless, the precise identities of human genes that confer vulnerability to problem drug use remain mostly unknown. Here, we describe a natural single nucleotide polymorphism in the human gene that encodes the principal endocannabinoid-inactivating enzyme, fatty acid amide hydrolase (FAAH), that in homozygous form is strongly associated with both street drug use and problem drug/alcohol use. This single nucleotide polymorphism results in a missense mutation (385C-->A) that converts a conserved proline residue to threonine (Pro129-->Thr), producing a FAAH variant that displays normal catalytic properties but an enhanced sensitivity to proteolytic degradation. Collectively, these results suggest that genetic mutations in FAAH may constitute important risk factors for problem drug use and support a potential link between functional abnormalities in the endogenous cannabinoid system and drug abuse and dependence.


Subject(s)
Amidohydrolases/genetics , Mutation, Missense , Substance-Related Disorders/genetics , Alcohol Drinking/genetics , Amidohydrolases/chemistry , Amidohydrolases/metabolism , Animals , Cannabinoid Receptor Modulators , Catalysis , Circular Dichroism , Genetic Predisposition to Disease , Genetic Variation , Genotype , Humans , Illicit Drugs , Odds Ratio , Polymerase Chain Reaction , Rats , Substance-Related Disorders/enzymology , Surveys and Questionnaires , Urea/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...