Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 422
Filter
Add more filters

Publication year range
1.
Org Biomol Chem ; 14(2): 612-622, 2016 Jan 14.
Article in English | MEDLINE | ID: mdl-26537174

ABSTRACT

α-Methylacyl-CoA racemase (AMACR; P504S) catalyses a key step in the degradation of branched-chain fatty acids and is important for the pharmacological activation of Ibuprofen and related drugs. Levels of AMACR are increased in prostate and other cancers, and it is a drug target. Development of AMACR as a drug target is hampered by lack of a convenient assay. AMACR irreversibly catalyses the elimination of HF from 3-fluoro-2-methylacyl-CoA substrates, and this reaction was investigated for use as an assay. Several known inhibitors and alternative substrates reduced conversion of 3-fluoro-2-methyldecanoyl-CoA by AMACR, as determined by (1)H NMR. The greatest reduction of activity was observed with known potent inhibitors. A series of novel acyl-CoA esters with aromatic side chains were synthesised for testing as chromophoric substrates. These acyl-CoA esters were converted to unsaturated products by AMACR, but their use was limited by non-enzymatic elimination. Fluoride sensors were also investigated as a method of quantifying released fluoride and thus AMACR activity. These sensors generally suffered from high background signal and lacked reproducibility under the assay conditions. In summary, the elimination reaction can be used to characterise inhibitors, but it was not possible to develop a convenient colorimetric or fluorescent assay using 3-fluoro-2-methylacyl-CoA substrates.


Subject(s)
Acyl Coenzyme A/pharmacology , Drug Evaluation, Preclinical , Enzyme Assays , Enzyme Inhibitors/pharmacology , Esters/pharmacology , Racemases and Epimerases/antagonists & inhibitors , Racemases and Epimerases/metabolism , Acyl Coenzyme A/chemical synthesis , Acyl Coenzyme A/chemistry , Biocatalysis , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Esters/chemical synthesis , Esters/chemistry , Humans , Molecular Structure , Racemases and Epimerases/chemistry , Structure-Activity Relationship
2.
Biochim Biophys Acta ; 1842(12 Pt A): 2510-6, 2014 Dec.
Article in English | MEDLINE | ID: mdl-23643712

ABSTRACT

Hyperammonemia is a frequent finding in various organic acidemias. One possible mechanism involves the inhibition of the enzyme N-acetylglutamate synthase (NAGS), by short-chain acyl-CoAs which accumulate due to defective catabolism of amino acids and/or fatty acids in the cell. The aim of this study was to investigate the effect of various acyl-CoAs on the activity of NAGS in conjunction with the formation of glutamate esters. NAGS activity was measured in vitro using a sensitive enzyme assay with ultraperformance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) product analysis. Propionyl-CoA and butyryl-CoA proved to be the most powerful inhibitors of N-acetylglutamate (NAG) formation. Branched-chain amino acid related CoAs (isovaleryl-CoA, 3-methylcrotonyl-CoA, isobutyryl-CoA) showed less pronounced inhibition of NAGS whereas the dicarboxylic short-chain acyl-CoAs (methylmalonyl-CoA, succinyl-CoA, glutaryl-CoA) had the least inhibitory effect. Subsequent work showed that the most powerful inhibitors also proved to be the best substrates in the formation of N-acylglutamates. Furthermore, we identified N-isovalerylglutamate, N-3-methylcrotonylglutamate and N-isobutyrylglutamate (the latter two in trace amounts), in the urines of patients with different organic acidemias. Collectively, these findings explain one of the contributing factors to secondary hyperammonemia, which lead to the reduced in vivo flux through the urea cycle in organic acidemias and result in the inadequate elimination of ammonia.


Subject(s)
Acyl Coenzyme A/pharmacology , Amino-Acid N-Acetyltransferase/antagonists & inhibitors , Amino-Acid N-Acetyltransferase/metabolism , Glutamic Acid/metabolism , Acyl Coenzyme A/metabolism , Carboxylic Acids/metabolism , Chromatography, High Pressure Liquid/methods , Dicarboxylic Acids/metabolism , Dose-Response Relationship, Drug , Esters , Glutamic Acid/chemistry , Humans , Hyperammonemia/metabolism , Kinetics , Tandem Mass Spectrometry
3.
J Inherit Metab Dis ; 37(3): 353-7, 2014 May.
Article in English | MEDLINE | ID: mdl-24154984

ABSTRACT

BACKGROUND: Valproic acid (VPA) is an effective antiepileptic drug that may induce progressive microvesicular steatosis. The impairment of mitochondrial function may be an important metabolic effect of VPA treatment with potential adverse consequences. OBJECTIVE: To investigate the influence of VPA on the activity of GTP- and ATP-specific succinate:CoA ligases (G-SUCL and A-SUCL). METHODS: The GTP- and ATP-specific SUCL activities were measured in human fibroblasts in the reverse direction, i.e. the formation of succinyl-CoA. These were assessed at different concentrations of succinate in the presence of VPA, valproyl-CoA and zinc chloride, an established inhibitor of the enzymes. Activities were measured using an optimized HPLC procedure. RESULTS: Valproyl-CoA (1 mM) inhibited the activity of A-SUCL and G-SUCL by 45-55% and 25-50%, respectively. VPA (1 mM) had no influence on the activity of the two enzymes. DISCUSSION: Valproyl-CoA appears to affect the activity of SUCL, especially with the ATP-specific enzyme. Considering the key role of SUCL in the Krebs cycle, interference with its activity might impair the cellular energy status. Moreover, A-SUCL is bound to the nucleoside diphosphate kinase (NDPK), which is responsible for the mitochondrial (deoxy)nucleotide synthesis. An inhibition of A-SUCL might influence the activity of NDPK inducing an imbalance of nucleotides in the mitochondria and eventually mitochondrial DNA depletion. This may account for the potential liver failure associated with valproate therapy, reported in patients with deficiencies within the mitochondrial DNA replicase system such as polymerase gamma 1.


Subject(s)
Acyl Coenzyme A/pharmacology , Adenosine Triphosphate/physiology , Guanosine Triphosphate/physiology , Succinate-CoA Ligases/antagonists & inhibitors , DNA, Mitochondrial/metabolism , Humans , Liver Failure/chemically induced , Nucleoside-Diphosphate Kinase/physiology , Valproic Acid/adverse effects , Valproic Acid/pharmacology
4.
Drug Metab Pharmacokinet ; 52: 100509, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37515836

ABSTRACT

Although acyl-CoA conjugates are known to have higher reactivity than acyl glucuronides, few studies have been conducted to evaluate the risk of the conjugates. In the present study, we aimed to develop a trapping assay for acyl-CoA conjugates using trapping reagents we have developed previously. It was revealed that Cys-Dan, which has both a thiol and an amino group, was the most effective in forming stable adducts containing an amide bond after intramolecular acyl migration. Additionally, we also developed a hepatocyte-based trapping assay in the present study to overcome the shortcomings of liver microsomes. Although liver microsomes are commonly used as enzyme sources in trapping assays, they lack some of the enzymes required for drug metabolism and detoxification systems. In human hepatocytes, our three trapping reagents, CysGlu-Dan, Dap-Dan and Cys-Dan, captured CYP-dependent reactive metabolites, reactive acyl glucuronides, and reactive acyl-CoA conjugates, respectively. The work suggests that the trapping assay with the reagents in hepatocytes is useful to evaluate the risk of reactive metabolites in drug discovery.


Subject(s)
Acyl Coenzyme A , Glucuronides , Humans , Acyl Coenzyme A/chemistry , Acyl Coenzyme A/metabolism , Acyl Coenzyme A/pharmacology , Indicators and Reagents/metabolism , Glucuronides/metabolism , Microsomes, Liver/metabolism , Sulfhydryl Compounds/metabolism , Sulfhydryl Compounds/pharmacology
5.
Life Sci ; 333: 122157, 2023 Nov 15.
Article in English | MEDLINE | ID: mdl-37805164

ABSTRACT

AIMS: This study aimed to investigate the effect and mechanism of methylcrotonyl-CoA carboxylase subunit 1 (MCCA) on multidrug resistance in multiple myeloma (MM). MATERIALS AND METHODS: The apoptosis kit and CCK-8 reagent were used to detect drug-induced cell apoptosis and viability. Immunoprecipitation, immunofluorescence staining, and protein structural simulation were used to detect the interaction between MCCA and Bad. Immunodeficient mice were injected with ARD cells and treated with bortezomib. Changes in tumor burden were recorded by bioluminescence imaging, and κ light chain content in the blood of mice was detected by enzyme-linked immunoassay. KEY FINDINGS: Patients with high MCCA expression from a primary MM dataset had superior overall survival. After treatment with different anti-MM drugs, MCCA knockdown MM (MCCA-KD) cells had higher survival rates than control knockdown (CTR-KD) cells (p < 0.05). Mechanistic studies have revealed that MCCA-KD cells had dysfunctional mitochondria with decreased Bax and Bad levels and increased Bcl-xl and Mcl-1 levels. Furthermore, that MCCA and Bad demonstrated protein-protein interactions. The half-life of Bad in MCCA-KD cells is significantly shorter than that in CTR-KD cells (7.34 vs. 2.42 h, p < 0.05). In a human MM xenograft mouse model, we confirmed that MCCA-KD tumors had a poor response to anti-MM drugs in vivo. Finally, we showed that MCCA might contribute to multidrug resistance in different human cancers, particularly in solid tumors. SIGNIFICANCE: Our findings demonstrated a novel function of MCCA in multidrug resistance. The lack of MCCA expression promoted antiapoptotic cell signaling in MM cells.


Subject(s)
Multiple Myeloma , Humans , Animals , Mice , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , Acyl Coenzyme A/pharmacology , Acyl Coenzyme A/therapeutic use , Bortezomib/pharmacology , Apoptosis , Cell Line, Tumor , Drug Resistance, Multiple , Drug Resistance, Neoplasm
6.
Differentiation ; 82(4-5): 244-52, 2011.
Article in English | MEDLINE | ID: mdl-21862201

ABSTRACT

The origins of benign prostatic diseases, such as benign prostatic hyperplasia (BPH) and chronic prostatitis/chronic pelvic pain syndrome (CP/CPPS), are poorly understood. Patients suffering from benign prostatic symptoms report a substantially reduced quality of life, and the relationship between benign prostate conditions and prostate cancer is uncertain. Epidemiologic data for BPH and CP/CPPS are limited, however an apparent association between BPH symptoms and cardiovascular disease (CVD) has been consistently reported. The prostate synthesizes and stores large amounts of cholesterol and prostate tissues may be particularly sensitive to perturbations in cholesterol metabolism. Hypercholesterolemia, a major risk factor for CVD, is also a risk factor for BPH. Animal model and clinical trial findings suggest that agents that inhibit cholesterol absorption from the intestine, such as the class of compounds known as polyene macrolides, can reduce prostate gland size and improve lower urinary tract symptoms (LUTS). Observational studies indicate that cholesterol-lowering drugs reduce the risk of aggressive prostate cancer, while prostate cancer cell growth and survival pathways depend in part on cholesterol-sensitive biochemical mechanisms. Here we review the evidence that cholesterol metabolism plays a role in the incidence of benign prostate disease and we highlight possible therapeutic approaches based on this concept.


Subject(s)
Cardiovascular Diseases/metabolism , Cholesterol/metabolism , Prostate/metabolism , Prostatic Hyperplasia/metabolism , Prostatic Neoplasms/metabolism , Prostatitis/metabolism , Acyl Coenzyme A/pharmacology , Anticholesteremic Agents/pharmacology , Cardiovascular Diseases/complications , Cardiovascular Diseases/epidemiology , Cholesterol/blood , Clinical Trials as Topic , Gene Expression , Humans , Hypercholesterolemia/complications , Hypercholesterolemia/metabolism , Macrolides/therapeutic use , Male , Prostate/drug effects , Prostate/pathology , Prostatic Hyperplasia/complications , Prostatic Hyperplasia/drug therapy , Prostatic Hyperplasia/epidemiology , Prostatic Neoplasms/complications , Prostatic Neoplasms/epidemiology , Prostatitis/complications , Prostatitis/epidemiology , Signal Transduction
7.
Biophys J ; 100(3): 620-628, 2011 Feb 02.
Article in English | MEDLINE | ID: mdl-21281576

ABSTRACT

The lipid bilayer is a critical determinant of ion channel activity; however, efforts to define the lipid dependence of channel function have generally been limited to cellular expression systems in which the membrane composition cannot be fully controlled. We reconstituted purified human Kir2.1 and Kir2.2 channels into liposomes of defined composition to study their phospholipid dependence of activity using (86)Rb(+) flux and patch-clamp assays. Our results demonstrate that Kir2.1 and Kir2.2 have two distinct lipid requirements for activity: a specific requirement for phosphatidylinositol 4,5-bisphosphate (PIP(2)) and a nonspecific requirement for anionic phospholipids. Whereas we previously showed that PIP(2) increases the channel open probability, in this work we find that activation by POPG increases both the open probability and unitary conductance. Oleoyl CoA potently inhibits Kir2.1 by antagonizing the specific requirement for PIP(2), and EPC appears to antagonize activation by the nonspecific anionic requirement. Phosphatidylinositol phosphates can act on both lipid requirements, yielding variable and even opposite effects on Kir2.1 activity depending on the lipid background. Mutagenesis experiments point to the role of intracellular residues in activation by both PIP(2) and anionic phospholipids. In conclusion, we utilized purified proteins in defined lipid membranes to quantitatively determine the phospholipid requirements for human Kir channel activity.


Subject(s)
Phospholipids/metabolism , Potassium Channels, Inwardly Rectifying/metabolism , Acyl Coenzyme A/pharmacology , Amino Acids/metabolism , Animals , Anions , Cattle , Humans , Intracellular Space/drug effects , Intracellular Space/metabolism , Ion Channel Gating/drug effects , Phosphatidylcholines/pharmacology , Phosphatidylglycerols/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Potassium Channels, Inwardly Rectifying/antagonists & inhibitors
8.
Pflugers Arch ; 461(6): 635-43, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21360038

ABSTRACT

Connexins form hemichannels at undocked plasma membranes and gap-junction channels (GJCs) at intercellular contacting zones. Under physiological conditions, hemichannels have low open probabilities, but their activation under pathological conditions, such as ischemia, induces and/or accelerates cell death. Connexin 46 (Cx46) is a major connexin of the lens, and mutations of this connexin induce cataracts. Here, we report the effects of linoleic acid (LA) on the electrical properties of Cx46 GJCs and hemichannels expressed in Xenopus laevis oocytes. LA has a biphasic effect, increasing hemichannel current at 0.1 µM and decreasing it at concentrations of 100 µM or higher. The effects of extracellular and microinjected LA conjugated to coenzyme A (LA-CoA) suggest that the current activation site is accessible from the intracellular but not extracellular compartment, whereas the current inhibitory site is either located in a region of the hemichannel pore inaccessible to intracellular LA-CoA, or requires crossing of LA through an organelle membrane. Experiments with other fatty acids demonstrated that the block of hemichannels depends on the presence of a hydrogenated double bond at position 9 and is directly proportional to the number of double bonds. Experiments in paired oocytes expressing Cx46 showed that LA does not affect GJCs. The block by unsaturated fatty acids reported here opens the possibility that increases in the concentration of these lipids in the lens induce cataract formation by blocking Cx46 hemichannels.


Subject(s)
Connexins/physiology , Gap Junctions/drug effects , Ion Channels/drug effects , Linoleic Acid/pharmacology , Acyl Coenzyme A/metabolism , Acyl Coenzyme A/pharmacology , Animals , Arachidonic Acid/pharmacology , Calcium/metabolism , Cataract/etiology , Fatty Acids, Unsaturated/pharmacology , Gap Junctions/physiology , Ion Channels/physiology , Lens, Crystalline/drug effects , Lens, Crystalline/physiopathology , Naphthalenes/pharmacology , Oocytes/drug effects , Protein Kinase C/physiology , Xenopus laevis
9.
Biochemistry ; 49(48): 10319-28, 2010 Dec 07.
Article in English | MEDLINE | ID: mdl-20977214

ABSTRACT

Catalysis by succinyl-CoA:3-oxoacid CoA transferase proceeds through a thioester intermediate in which CoA is covalently linked to the enzyme. To determine the conformation of the thioester intermediate, crystals of the pig enzyme were grown in the presence of the substrate acetoacetyl-CoA. X-ray diffraction data show the enzyme in both the free form and covalently bound to CoA via Glu305. In the complex, the protein adopts a conformation in which residues 267-275, 280-287, 357-373, and 398-477 have shifted toward Glu305, closing the enzyme around the thioester. Enzymes provide catalysis by stabilizing the transition state relative to complexes with substrates or products. In this case, the conformational change allows the enzyme to interact with parts of CoA distant from the reactive thiol while the thiol is covalently linked to the enzyme. The enzyme forms stabilizing interactions with both the nucleotide and pantoic acid portions of CoA, while the interactions with the amide groups of the pantetheine portion are poor. The results shed light on how the enzyme uses the binding energy for groups remote from the active center of CoA to destabilize atoms closer to the active center, leading to acceleration of the reaction by the enzyme.


Subject(s)
Acyl Coenzyme A/metabolism , Acyl Coenzyme A/pharmacology , Biocatalysis , Coenzyme A-Transferases/chemistry , Coenzyme A-Transferases/metabolism , Animals , Binding Sites , Crystallography, X-Ray , Enzyme Stability/drug effects , Kinetics , Models, Molecular , Protein Binding , Protein Conformation/drug effects , Swine
10.
Antiviral Res ; 177: 104734, 2020 05.
Article in English | MEDLINE | ID: mdl-32057770

ABSTRACT

Hepatitis C virus (HCV) is a leading cause of chronic hepatitis and end-stage liver diseases. Mature HCV virions are bound by host-derived lipoproteins. Lack of an HCV vaccine warrants a major role of antiviral treatment in the global elimination of hepatitis C. Although direct-acting antivirals (DAAs) are replacing the interferon-based treatment and have dramatically improved the cure rate, the presence of viral variants resistant to DAAs, HCV genotype/subtype-specific efficacy, and high cost of DAAs argue novel and affordable regimens. In this study, we identified the antiviral effects of long-chain fatty acyl-coenzyme A (LCFA-CoA) against the infections of HCV genotypes 1-6 through targeting mature HCV-bound lipoproteins, suggesting novel mechanism(s) of antiviral different from those used by host-targeting agents or DAAs. We found that the antiviral activity of LCFA-CoA relied on the long-chain saturated fatty acid and the CoA group, and was enhanced when combined with pegylated-interferon or DAAs. Importantly, we demonstrated that LCFA-CoA efficiently inhibited the infection of HCV variants carrying DAA-resistant mutations. The mechanistic study revealed that LCFA-CoA specifically abolished the attachment and binding steps and also inhibited the cell-to-cell viral transmission. LCFA-CoA targeted mature HCV-bound lipoproteins, but not apolipoproteins B or E. In addition, LCFA-CoA could also inhibit the infection of the dengue virus. Our findings suggest that LCFA-CoA could potentially serve as a supplement HCV therapy, particularly for the DAA-resistant HCV variants. Taken together, LCFA-CoA may be further developed to be a novel class of antivirals with mechanism(s), different from host-targeting agents or DAAs, of targeting the components associated with mature HCV virions.


Subject(s)
Acyl Coenzyme A/pharmacology , Antiviral Agents/pharmacology , Hepacivirus/drug effects , Lipoproteins/antagonists & inhibitors , Virus Internalization/drug effects , Cell Line, Tumor , Genotype , Hepacivirus/genetics , Humans , Virion/drug effects
11.
Biochem Biophys Res Commun ; 381(4): 496-501, 2009 Apr 17.
Article in English | MEDLINE | ID: mdl-19233137

ABSTRACT

ATP-sensitive K(+) (K(ATP)) channels couple intermediary metabolism to cellular activity. Genetic disruption of these channels impairs glucose homeostasis. Similar effects occur from a single-nucleotide polymorphism of the Kir6.2 subunit seen in greater than 50% of the human population, which causes a point mutation of Glu23 to lysine. This E23K variant shows higher susceptibility to diabetes due to mechanisms that are not fully understood. This study was designed to examine the dysregulation of E23K on insulin sensitivity in the presence of long-chain fatty acyl CoA (LC-CoA), a major active form of free fatty acids. Physiological concentrations of LC-CoA decreased insulin sensitivity in E23K-transfected L6 muscle cells by increasing the activation of negative regulators in the insulin signaling pathway. LC-CoA also reduced IRS-1 and Akt phosphorylation and glucose transport. This effect was not due to the expression of the E23K mutant on cell membrane. Our results indicate that E23K could impair insulin sensitivity, thus predisposing E23K carriers to insulin resistance.


Subject(s)
Acyl Coenzyme A/metabolism , Insulin Resistance/genetics , Insulin/metabolism , Muscle, Skeletal/drug effects , Potassium Channels, Inwardly Rectifying/genetics , Acyl Coenzyme A/pharmacology , Animals , Cell Line , Glutamic Acid/genetics , Glutamic Acid/metabolism , Humans , Insulin/pharmacology , Lysine/genetics , Lysine/metabolism , Muscle, Skeletal/metabolism , Polymorphism, Genetic , Rats
12.
J Cell Biol ; 110(4): 955-61, 1990 Apr.
Article in English | MEDLINE | ID: mdl-2324202

ABSTRACT

Two different methods, stimulation of transport by fatty acyl-coenzyme A (CoA) and inhibition of transport by a nonhydrolyzable analogue of palmitoyl-CoA, reveal that fatty acylation is required to promote fusion of transport vesicles with Golgi cisternae. Specifically, fatty acyl-CoA is needed after the attachment of coated vesicles and subsequent uncoating of the vesicles, and after the binding of the NEM-sensitive fusion protein (NSF) to the membranes, but before the actual fusion event. We therefore suggest that an acylated transport component participates, directly or indirectly, in membrane fusion.


Subject(s)
Acyl Coenzyme A/pharmacology , Carrier Proteins/metabolism , Golgi Apparatus/metabolism , Intracellular Membranes/metabolism , Membrane Fusion/drug effects , Organelles/metabolism , Vesicular Transport Proteins , Biological Transport , Cell-Free System , Coenzyme A/pharmacology , Detergents/pharmacology , Ethanol/pharmacology , Golgi Apparatus/drug effects , Intracellular Membranes/drug effects , Kinetics , N-Ethylmaleimide-Sensitive Proteins , Octoxynol , Organelles/drug effects , Polyethylene Glycols/pharmacology
13.
Appl Environ Microbiol ; 74(10): 3229-41, 2008 May.
Article in English | MEDLINE | ID: mdl-18344344

ABSTRACT

Producing complex chemicals using synthetic metabolic pathways in microbial hosts can have many advantages over chemical synthesis but is often complicated by deleterious interactions between pathway intermediates and the host cell metabolism. With the maturation of functional genomic analysis, it is now technically feasible to identify modes of toxicity associated with the accumulation of foreign molecules in the engineered bacterium. Previously, Escherichia coli was engineered to produce large quantities of isoprenoids by creating a mevalonate-based isopentenyl pyrophosphate biosynthetic pathway (V. J. J. Martin et al., Nat. Biotechnol. 21:796-802, 2003). The engineered E. coli strain produced high levels of isoprenoids, but further optimization led to an imbalance in carbon flux and the accumulation of the pathway intermediate 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA), which proved to be cytotoxic to E. coli. Using both DNA microarray analysis and targeted metabolite profiling, we have studied E. coli strains inhibited by the intracellular accumulation of HMG-CoA. Our results indicate that HMG-CoA inhibits fatty acid biosynthesis in the microbial host, leading to generalized membrane stress. The cytotoxic effects of HMG-CoA accumulation can be counteracted by the addition of palmitic acid (16:0) and, to a lesser extent, oleic acid (cis-Delta(9)-18:1) in the growth medium. This work demonstrates the utility of using transcriptomic and metabolomic methods to optimize synthetic biological systems.


Subject(s)
Escherichia coli/genetics , Escherichia coli/metabolism , Metabolic Networks and Pathways/genetics , Terpenes/metabolism , Acyl Coenzyme A/analysis , Acyl Coenzyme A/pharmacology , Anti-Bacterial Agents/analysis , Anti-Bacterial Agents/pharmacology , Cell Membrane/physiology , Escherichia coli/chemistry , Escherichia coli/growth & development , Fatty Acids/antagonists & inhibitors , Fatty Acids/biosynthesis , Gene Expression Profiling , Oleic Acid/metabolism , Palmitic Acid/metabolism
14.
Org Lett ; 10(11): 2235-8, 2008 Jun 05.
Article in English | MEDLINE | ID: mdl-18442241

ABSTRACT

Oct-2-yn-4-enoyl-CoA was found to be a multifunctional irreversible enzyme inhibitor in fatty acid oxidation mainly targeting mitochondrial trifunctional protein beta-subunit. It can also inactivate enoyl-CoA hydratase 2 and medium-chain acyl-CoA dehydrogenase. This study increased our understanding for the effect of acetylenic acids on fatty acid oxidation.


Subject(s)
Acyl Coenzyme A/chemistry , Acyl Coenzyme A/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Fatty Acids/chemistry , Acyl-CoA Dehydrogenase/antagonists & inhibitors , Acyl-CoA Dehydrogenase/chemistry , Amino Acid Sequence , Animals , Enoyl-CoA Hydratase/antagonists & inhibitors , Enoyl-CoA Hydratase/chemistry , Mitochondrial Trifunctional Protein , Molecular Sequence Data , Multienzyme Complexes/antagonists & inhibitors , Multienzyme Complexes/chemistry , Oxidation-Reduction/drug effects , Rats
15.
Org Lett ; 10(15): 3355-8, 2008 Aug 07.
Article in English | MEDLINE | ID: mdl-18611036

ABSTRACT

Enoyl-CoA hydratase 1 and enoyl-CoA hydratase 2 in long-chain fatty acid oxidation were comparatively investigated through mechanistic studies for inactivation of the enzymes with methylenecyclopropylformyl-CoA and 3-octynoyl-CoA. Methylenecyclopropylformyl-CoA can inactivate both enzymes, while 3-octynoyl-CoA inactivates enoyl-CoA hydratase 2 only. The study increased our understanding of these two enzymes in fatty acid oxidation.


Subject(s)
Acyl Coenzyme A/chemistry , Enoyl-CoA Hydratase/antagonists & inhibitors , Enoyl-CoA Hydratase/metabolism , Fatty Acids/metabolism , Acyl Coenzyme A/pharmacology , Amino Acid Sequence , Animals , Enoyl-CoA Hydratase/chemistry , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Fatty Acids/chemistry , Isoenzymes/antagonists & inhibitors , Isoenzymes/chemistry , Isoenzymes/metabolism , Mitochondria/enzymology , Molecular Sequence Data , Oxidation-Reduction , Peroxisomes/chemistry , Peroxisomes/metabolism , Rats , Tandem Mass Spectrometry
16.
Article in English | MEDLINE | ID: mdl-18083017

ABSTRACT

The effects of palmitic acid (PA), stearic acid (SA) and oleic acid (OA), and their respective CoA esters, PA-CoA, SA-CoA and OA-CoA, on the activities of cyclooxygenase (COX)-1 and -2 were examined. Ten units of purified COX-1 or -2 were preincubated with drugs in the presence of hematin (0.1 microM) and phenol (2 mM) as cofactors for 10 min at 37 degrees C, and then incubated with 100 microM arachidonic acid for 2 min at 37 degrees C. The amounts of prostaglandins formed were measured by HPLC. PA, SA and OA had no effect on the COX-1 and -2 activities, but their respective CoA esters, PA-CoA, SA-CoA and OA-CoA, suppressed COX-1 activity with no significant effect on COX-2 activity. The inhibitory effect of SA-CoA was much stronger than that of PA-CoA and OA-CoA. These results suggest that SA has the potential to inhibit COX-1 activity, but not COX-2 activity, in the form of their CoA ester.


Subject(s)
Acyl Coenzyme A/pharmacology , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/metabolism , Stearic Acids/pharmacology , Animals , Arachidonic Acid/metabolism , Cattle , Oleic Acid/pharmacology , Palmitic Acid/pharmacology , Prostaglandins/metabolism
17.
Mol Cancer Ther ; 17(2): 407-418, 2018 02.
Article in English | MEDLINE | ID: mdl-29030460

ABSTRACT

Despite significant advances in combinations of radiotherapy and chemotherapy, altered fractionation schedules and image-guided radiotherapy, many cancer patients fail to benefit from radiation. A prevailing hypothesis is that targeting repair of DNA double strand breaks (DSB) can enhance radiation effects in the tumor and overcome therapeutic resistance without incurring off-target toxicities. Unrepaired DSBs can block cancer cell proliferation, promote cancer cell death, and induce cellular senescence. Given the slow progress to date translating novel DSB repair inhibitors as radiosensitizers, we have explored drug repurposing, a proven route to improving speed, costs, and success rates of drug development. In a prior screen where we tracked resolution of ionizing radiation-induced foci (IRIF) as a proxy for DSB repair, we had identified pitavastatin (Livalo), an HMG-CoA reductase inhibitor commonly used for lipid lowering, as a candidate radiosensitizer. Here, we report that pitavastatin and other lipophilic statins are potent inhibitors of DSB repair in breast and melanoma models both in vitro and in vivo When combined with ionizing radiation, pitavastatin increased persistent DSBs, induced senescence, and enhanced acute effects of radiation on radioresistant melanoma tumors. shRNA knockdown implicated HMG-CoA reductase, farnesyl diphosphate synthase, and protein farnesyl transferase in IRIF resolution, DSB repair, and senescence. These data confirm on-target activity of statins, although via inhibition of protein prenylation rather than cholesterol biosynthesis. In light of prior studies demonstrating enhanced efficacy of radiotherapy in patients taking statins, this work argues for clinical evaluation of lipophilic statins as nontoxic radiosensitizers to enhance the benefits of image-guided radiotherapy. Mol Cancer Ther; 17(2); 407-18. ©2017 AACRSee all articles in this MCT Focus section, "Developmental Therapeutics in Radiation Oncology."


Subject(s)
DNA Repair/drug effects , Acyl Coenzyme A/pharmacology , Animals , Cellular Senescence , Female , Humans , Mice
18.
J Clin Invest ; 64(6): 1544-51, 1979 Dec.
Article in English | MEDLINE | ID: mdl-500823

ABSTRACT

In the search for the mechanism by which hyperammonemia complicates propionic and methylmalonic acidemia the effects of a series of acyl-coenzyme A (CoA) derivatives were studied on the activity of N-acetylglutamate synthetase in rat liver mitochondria using acetyl-CoA as substrate. Propionyl-CoA was found to be a competitive inhibitor. The inhibition constant of 0.71 mM is in the range of concentrations of propionate found in the serum of patients with propionic and methylmalonic acidemia. Propionyl-CoA was also found to be a substrate for N-acetylglutamate synthetase, forming N-propionylglutamate. This compound was a weak activator of rat liver carbamoylphosphate synthetase; the activation constant was 1.1 mM as compared with 0.12 mM for N-acetylglutamate. A decreased level of N-acetylglutamate in liver mitochondria that would follow inhibition of N-acetylglutamate synthetase by propionyl-CoA would be expected to lead to hyperammonemia. Methylmalonyl-CoA, tiglyl-CoA, and isovaleryl-CoA at a concentration of 3 mM caused 30-70% inhibition of N-acetylglutamate synthetase. 3the latter two compounds are readily detoxified by the formation of N-acylglycine conjugates in liver, which may prevent large accumulations and could explain why hyperammonemia is not characteristic of patients with beta-ketothiolase deficiency or isovaleric acidemia in whom these compounds would be expected to be elevated.


Subject(s)
Acetyltransferases/antagonists & inhibitors , Acidosis/metabolism , Acyl Coenzyme A/pharmacology , Malonates/blood , Methylmalonic Acid/blood , Mitochondria, Liver/enzymology , Propionates/blood , Acetyl Coenzyme A , Ammonia/blood , Animals , Glutamates , Kinetics , Rats , Substrate Specificity
19.
J Med Chem ; 50(11): 2700-7, 2007 May 31.
Article in English | MEDLINE | ID: mdl-17477519

ABSTRACT

The enzyme alpha-methylacyl-CoA racemase (AMACR) is overexpressed in prostate, colon, and other cancers and has been partially validated as a potential therapeutic target by siRNA knockdown of the AMACR gene. Analogs of the natural substrate branched chain alpha-methylacyl coenzyme A esters, possessing one or more beta-fluorine atoms, have been synthesized using Wittig, conjugate addition, and asymmetric aldol reactions and found to be reversible competitive inhibitors. Each diastereomer of the previously reported inhibitor ibuprofenoyl-CoA was also tested. The compounds had Ki values of 0.9-20 microM and are the most potent inhibitors yet known. The presence of beta-fluorine on the alpha-methyl group or the acyl chain results in a significant lowering of the Ki value compared with nonfluorinated analogs, and this is attributed to a lowering of the pKa of the alpha-proton, facilitating enolization and binding. Several of the CoA ester inhibitors were formed by incubating the free carboxylic acid precursors with cell free extracts and CoA. alpha-Trifluoromethyltetradecanoic acid, the precursor to the most potent inhibitor, was shown to inhibit growth of cancer cell lines PC3, CWR22 Rv1, and Du145 in a dose-dependent manner and could be related to the expression level of AMACR.


Subject(s)
Acyl Coenzyme A/chemical synthesis , Antineoplastic Agents/chemical synthesis , Myristates/chemical synthesis , Racemases and Epimerases/antagonists & inhibitors , Acyl Coenzyme A/chemistry , Acyl Coenzyme A/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Binding Sites , Cell Proliferation/drug effects , Crystallography, X-Ray , Drug Design , Drug Screening Assays, Antitumor , Humans , Male , Myristates/chemistry , Myristates/pharmacology , Prostatic Neoplasms , Racemases and Epimerases/chemistry , Stereoisomerism
20.
Life Sci ; 80(14): 1282-91, 2007 Mar 13.
Article in English | MEDLINE | ID: mdl-17257628

ABSTRACT

Inducible nitric oxide synthase (iNOS) is an homodimeric enzyme which produces large amounts of nitric oxide (NO) in response to inflammatory stimuli. Several factors affect the synthesis and catalytic activity of iNOS. Particularly, dimerization of NOS monomers is promoted by heme, whereas an intracellular depletion of heme and/or L-arginine considerably decreases NOS resistance to proteolysis. In this study, we found that oxalomalate (OMA, oxalomalic acid, alpha-hydroxy-beta-oxalosuccinic acid), an inhibitor of both aconitase and NADP-dependent isocitrate dehydrogenase, inhibited nitrite production and iNOS protein expression in lipopolysaccharide (LPS)-activated J774 macrophages, without affecting iNOS mRNA content. Furthermore, injection of OMA precursors to LPS-stimulated rats also decreased nitrite production and iNOS expression in isolated peritoneal macrophages. Interestingly, alpha-ketoglutarate or succinyl-CoA administration reversed OMA effect on NO production, thus correlating NO biosynthesis with the anabolic capacity of Krebs cycle. When protein synthesis was blocked by cycloheximide in LPS-activated J774 cells treated with OMA, iNOS protein levels, evaluated by Western blot analysis and (35)S-metabolic labelling, were decreased, suggesting that OMA reduces iNOS biosynthesis and induces an increase in the degradation rate of iNOS protein. Moreover, we showed that OMA inhibits the activity of the iNOS from lung of LPS-treated rats by enzymatic assay. Our results, demonstrating that OMA acts regulating synthesis, catalytic activity and degradation of iNOS, suggest that this compound might have a potential role in reducing the NO overproduction occurring in some pathological conditions.


Subject(s)
Enzyme Inhibitors/pharmacology , Macrophages/drug effects , Nitric Oxide Synthase Type II/metabolism , Oxalates/pharmacology , Acyl Coenzyme A/pharmacology , Animals , Cell Line , Cell Survival/drug effects , Cycloheximide/pharmacology , Drug Antagonism , Gene Expression Regulation, Enzymologic/drug effects , Ketoglutaric Acids/pharmacology , Lipopolysaccharides/pharmacology , Lung/drug effects , Lung/enzymology , Macrophage Activation/drug effects , Macrophage Activation/immunology , Macrophages/enzymology , Male , Nitric Oxide Synthase Type II/genetics , Nitrites/metabolism , Protein Synthesis Inhibitors/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL