Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
J Biol Chem ; 299(12): 105366, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37863264

ABSTRACT

Hypoxic responses in plants involve Plant Cysteine Oxidases (PCOs). They catalyze the N-terminal cysteine oxidation of Ethylene Response Factors VII (ERF-VII) in an oxygen-dependent manner, leading to their degradation via the cysteine N-degron pathway (Cys-NDP) in normoxia. In hypoxia, PCO activity drops, leading to the stabilization of ERF-VIIs and subsequent hypoxic gene upregulation. Thus far, no chemicals have been described to specifically inhibit PCO enzymes. In this work, we devised an in vivo pipeline to discover Cys-NDP effector molecules. Budding yeast expressing AtPCO4 and plant-based ERF-VII reporters was deployed to screen a library of natural-like chemical scaffolds and was further combined with an Arabidopsis Cys-NDP reporter line. This strategy allowed us to identify three PCO inhibitors, two of which were shown to affect PCO activity in vitro. Application of these molecules to Arabidopsis seedlings led to an increase in ERF-VII stability, induction of anaerobic gene expression, and improvement of tolerance to anoxia. By combining a high-throughput heterologous platform and the plant model Arabidopsis, our synthetic pipeline provides a versatile system to study how the Cys-NDP is modulated. Its first application here led to the discovery of at least two hypoxia-mimicking molecules with the potential to impact plant tolerance to low oxygen stress.


Subject(s)
Arabidopsis Proteins , Cysteine Dioxygenase , Enzyme Inhibitors , Small Molecule Libraries , Humans , Arabidopsis/drug effects , Arabidopsis/metabolism , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Cysteine/metabolism , Cysteine Dioxygenase/antagonists & inhibitors , Cysteine Dioxygenase/metabolism , Gene Expression Regulation, Plant/drug effects , Oxygen/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Drug Evaluation, Preclinical/methods , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Seedlings/drug effects , Anaerobiosis , Degrons , Enzyme Activation/drug effects , Recombinant Proteins/metabolism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/isolation & purification , Enzyme Inhibitors/pharmacology
2.
Chemistry ; 26(6): 1328-1334, 2020 Jan 27.
Article in English | MEDLINE | ID: mdl-31545545

ABSTRACT

Sulfoxide synthases are non-heme iron enzymes that participate in the biosynthesis of thiohistidines, such as ergothioneine and ovothiol A. The sulfoxide synthase EgtB from Chloracidobacterium thermophilum (CthEgtB) catalyzes oxidative coupling between the side chains of N-α-trimethyl histidine (TMH) and cysteine (Cys) in a reaction that entails complete reduction of molecular oxygen, carbon-sulfur (C-S) and sulfur-oxygen (S-O) bond formation as well as carbon-hydrogen (C-H) bond cleavage. In this report, we show that CthEgtB and other bacterial sulfoxide synthases cannot efficiently accept selenocysteine (SeCys) as a substrate in place of cysteine. In contrast, the sulfoxide synthase from the filamentous fungus Chaetomium thermophilum (CthEgt1) catalyzes C-S and C-Se bond formation at almost equal efficiency. We discuss evidence suggesting that this functional difference between bacterial and fungal sulfoxide synthases emerges from different modes of oxygen activation.


Subject(s)
Acidobacteria/enzymology , Bacterial Proteins/antagonists & inhibitors , Fungal Proteins/antagonists & inhibitors , Selenocysteine/chemistry , Bacterial Proteins/metabolism , Binding Sites , Binding, Competitive , Biocatalysis , Catalytic Domain , Cysteine Dioxygenase/antagonists & inhibitors , Cysteine Dioxygenase/metabolism , Ergothioneine/chemistry , Ergothioneine/metabolism , Fungal Proteins/metabolism , Kinetics , Molecular Dynamics Simulation , Mycobacteriaceae/enzymology , Selenocysteine/metabolism
3.
J Biol Chem ; 289(44): 30800-30809, 2014 Oct 31.
Article in English | MEDLINE | ID: mdl-25228698

ABSTRACT

The versatile thiol mercaptosuccinate has a wide range of applications, e.g. in quantum dot research or in bioimaging. Its metabolism is investigated in Variovorax paradoxus strain B4, which can utilize this compound as the sole source of carbon and sulfur. Proteomic studies of strain B4 resulted in the identification of a putative mercaptosuccinate dioxygenase, a cysteine dioxygenase homologue, possibly representing the key enzyme in the degradation of mercaptosuccinate. Therefore, the putative mercaptosuccinate dioxygenase was heterologously expressed, purified, and characterized in this study. The results clearly demonstrated that the enzyme utilizes mercaptosuccinate with concomitant consumption of oxygen. Thus, the enzyme is designated as mercaptosuccinate dioxygenase. Succinate and sulfite were verified as the final reaction products. The enzyme showed an apparent Km of 0.4 mM, and a specific activity (Vmax) of 20.0 µmol min(-1) mg(-1) corresponding to a kcat of 7.7 s(-1). Furthermore, the enzyme was highly specific for mercaptosuccinate, no activity was observed with cysteine, dithiothreitol, 2-mercaptoethanol, and 3-mercaptopropionate. These structurally related thiols did not have an inhibitory effect either. Fe(II) could clearly be identified as metal cofactor of the mercaptosuccinate dioxygenase with a content of 0.6 mol of Fe(II)/mol of enzyme. The recently proposed hypothesis for the degradation pathway of mercaptosuccinate based on proteome analyses could be strengthened in the present study. (i) Mercaptosuccinate is first converted to sulfinosuccinate by this mercaptosuccinate dioxygenase; (ii) sulfinosuccinate is spontaneously desulfinated to succinate and sulfite; and (iii) whereas succinate enters the central metabolism, sulfite is detoxified by the previously identified putative molybdopterin oxidoreductase.


Subject(s)
Bacterial Proteins/chemistry , Comamonadaceae/enzymology , Cysteine Dioxygenase/chemistry , Thiomalates/chemistry , Amino Acid Sequence , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/isolation & purification , Cysteine Dioxygenase/antagonists & inhibitors , Cysteine Dioxygenase/isolation & purification , Enzyme Inhibitors/chemistry , Kinetics , Molecular Sequence Data , Substrate Specificity
4.
Anal Biochem ; 459: 56-60, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24857787

ABSTRACT

A fast and easy method for enzyme activity assays using the chromogenic Ellman reagent, 5,5'-dithiobis(2-nitrobenzoic acid), was developed. The method was used to measure the activity of the nonheme mono-iron enzyme cysteine dioxygenase. Quantifying the depletion of the substrate, cysteine, allowed standard kinetic parameters to be determined for the enzyme from Rattus norvegicus. The assay was also used to quickly test the effects of ionic strength, pH, enzyme storage conditions, and potential inhibitors and activators. This assay facilitates a higher throughput than available HPLC-based assays, as it enjoys the advantages of fewer sample handling steps, implementation in a 96-well format, and speed. In addition, the relative specificity of Ellman's reagent, coupled with its reaction with a wide range of thiols, means that this assay is applicable to many enzymes. Finally, the use of readily available reagents and instrumentation means that this assay can be used by practically any research group to compare results with those of other groups.


Subject(s)
Chromogenic Compounds/chemistry , Cysteine Dioxygenase/metabolism , Dithionitrobenzoic Acid/chemistry , Enzyme Assays/methods , Animals , Cysteine/metabolism , Cysteine Dioxygenase/antagonists & inhibitors , Enzyme Activators/pharmacology , Enzyme Inhibitors/pharmacology , Rats , Substrate Specificity
5.
Amino Acids ; 46(5): 1275-83, 2014 May.
Article in English | MEDLINE | ID: mdl-24553827

ABSTRACT

Taurine deficiency has been suggested to contribute to the pathogenesis and complications of advanced hepatic diseases. The molecular basis for a low level of taurine associated with hepatic failure is largely unknown. Using carbon tetrachloride (CCl4)-induced cirrhotic rat model, we found that the activity and expression of cysteine dioxygenase (CDO), a rate-limiting enzyme in taurine synthesis, were significantly decreased in the liver of these rats. To investigate the underlying mechanisms for the suppression, we examined the effects of pathological cytokines on CDO expression in human hepatoma HepG2 cells. Among the several cytokines, transforming growth factor-ß (TGF-ß), one of the key mediators of fibrogenesis, suppressed Cdo1 gene transcription through the MEK/ERK pathway. Finally, we further examined potential effects of branched-chain amino acids (BCAA) on CDO expression, as it has been reported that oral BCAA supplementation increased plasma taurine level in the patients with liver cirrhosis. BCAA, especially leucine, promoted Cdo1 gene transcription, and attenuated TGF-ß-mediated suppression of Cdo1 gene expression. These results indicate that the low plasma level of taurine in advanced hepatic disease is due to decreased hepatic CDO expression, which can be partly attributed to suppressive effect of TGF-ß on Cdo1 gene transcription. Furthermore, our observation that BCAA promotes Cdo1 expression suggests that BCAA may be therapeutically useful to improve hepatic taurine metabolism and further suppress dysfunctions associated with low level of taurine in hepatic diseases.


Subject(s)
Amino Acids, Branched-Chain/metabolism , Cysteine Dioxygenase/antagonists & inhibitors , Cysteine Dioxygenase/metabolism , Liver Cirrhosis/enzymology , Taurine/biosynthesis , Transforming Growth Factor beta1/metabolism , Animals , Cysteine Dioxygenase/genetics , Down-Regulation , Hep G2 Cells , Humans , Liver Cirrhosis/genetics , Liver Cirrhosis/metabolism , Male , Rats , Rats, Sprague-Dawley
6.
Cancer Res ; 74(3): 787-96, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24351290

ABSTRACT

The relevance of cysteine metabolism in cancer has gained considerable interest in recent years, largely focusing on its role in generating the antioxidant glutathione. Through metabolomic profiling using a combination of high-throughput liquid and gas chromatography-based mass spectrometry on a total of 69 patient-derived glioma specimens, this report documents the discovery of a parallel pathway involving cysteine catabolism that results in the accumulation of cysteine sulfinic acid (CSA) in glioblastoma. These studies identified CSA to rank as one of the top metabolites differentiating glioblastoma from low-grade glioma. There was strong intratumoral concordance of CSA levels with expression of its biosynthetic enzyme cysteine dioxygenase 1 (CDO1). Studies designed to determine the biologic consequence of this metabolic pathway identified its capacity to inhibit oxidative phosphorylation in glioblastoma cells, which was determined by decreased cellular respiration, decreased ATP production, and increased mitochondrial membrane potential following pathway activation. CSA-induced attenuation of oxidative phosphorylation was attributed to inhibition of the regulatory enzyme pyruvate dehydrogenase. Studies performed in vivo abrogating the CDO1/CSA axis using a lentiviral-mediated short hairpin RNA approach resulted in significant tumor growth inhibition in a glioblastoma mouse model, supporting the potential for this metabolic pathway to serve as a therapeutic target. Collectively, we identified a novel, targetable metabolic pathway involving cysteine catabolism contributing to the growth of aggressive high-grade gliomas. These findings serve as a framework for future investigations designed to more comprehensively determine the clinical application of this metabolic pathway and its contributory role in tumorigenesis.


Subject(s)
Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cysteine/metabolism , Glioblastoma/metabolism , Glioblastoma/pathology , Metabolic Networks and Pathways , Animals , Brain Neoplasms/genetics , Cell Line, Tumor , Cysteine/analogs & derivatives , Cysteine/pharmacology , Cysteine Dioxygenase/antagonists & inhibitors , Cysteine Dioxygenase/genetics , Cysteine Dioxygenase/metabolism , Disease Models, Animal , Enzyme Activation/drug effects , Gene Expression , Glioblastoma/genetics , Humans , Mice , Mice, Knockout , Mitochondria/drug effects , Mitochondria/metabolism , Neoplasm Grading , Pyruvate Dehydrogenase Complex/metabolism , Tumor Burden/drug effects , Tumor Burden/genetics
7.
Biochemistry ; 52(51): 9104-19, 2013 Dec 23.
Article in English | MEDLINE | ID: mdl-24279989

ABSTRACT

Cysteine dioxygenase (CDO) is a non-heme iron enzyme that catalyzes the O2-dependent oxidation of l-cysteine (l-Cys) to produce cysteinesulfinic acid (CSA). Adjacent to the Fe site of CDO is a covalently cross-linked cysteine-tyrosine pair (C93-Y157). While several theories have been proposed for the function of the C93-Y157 pair, the role of this post-translational modification remains unclear. In this work, the steady-state kinetics and O2/CSA coupling efficiency were measured for wild-type CDO and selected active site variants (Y157F, C93A, and H155A) to probe the influence of second-sphere enzyme-substrate interactions on catalysis. In these experiments, it was observed that both kcat and the O2/CSA coupling efficiency were highly sensitive to the presence of the C93-Y157 cross-link and its proximity to the substrate carboxylate group. Complementary electron paramagnetic resonance (EPR) experiments were performed to obtain a more detailed understanding of the second-sphere interactions identified in O2/CSA coupling experiments. Samples of the catalytically inactive substrate-bound Fe(III)-CDO species were treated with cyanide, resulting in a low-spin (S = ¹/2) ternary complex. Remarkably, both the presence of the C93-Y157 pair and interactions with the Cys carboxylate group could be readily identified by perturbations to the rhombic EPR signal. Spectroscopically validated active site quantum mechanics/molecular mechanics and density functional theory computational models are provided to suggest a potential role for Y157 in the positioning of the substrate Cys in the active site and to verify the orientation of the g-tensor relative to the CDO Fe site molecular axis.


Subject(s)
Cysteine Dioxygenase/metabolism , Iron/metabolism , Models, Molecular , Oxygen/metabolism , Protein Processing, Post-Translational , Amino Acid Substitution , Animals , Apoenzymes/chemistry , Apoenzymes/genetics , Apoenzymes/metabolism , Binding Sites , Biocatalysis/drug effects , Catalytic Domain , Cysteine/analogs & derivatives , Cysteine/chemistry , Cysteine/metabolism , Cysteine Dioxygenase/antagonists & inhibitors , Cysteine Dioxygenase/chemistry , Cysteine Dioxygenase/genetics , Enzyme Inhibitors/pharmacology , Holoenzymes/chemistry , Holoenzymes/genetics , Holoenzymes/metabolism , Kinetics , Mice , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Oxidation-Reduction/drug effects , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Tyrosine/chemistry , Tyrosine/metabolism
8.
Clin Cancer Res ; 19(12): 3201-11, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23630167

ABSTRACT

PURPOSE: Genome-wide DNA methylation analyses have identified hundreds of candidate DNA-hypermethylated genes in cancer. Comprehensive functional analyses provide an understanding of the biologic significance of this vast amount of DNA methylation data that may allow the determination of key epigenetic events associated with tumorigenesis. EXPERIMENTAL DESIGN: To study mechanisms of cysteine dioxygenase type 1 (CDO1) inactivation and its functional significance in breast cancer in a comprehensive manner, we screened for DNA methylation and gene mutations in primary breast cancers and analyzed growth, survival, and reactive oxygen species (ROS) production in breast cancer cells with restored CDO1 function in the context of anthracycline treatment. RESULTS: DNA methylation-associated silencing of CDO1 in breast cancer is frequent (60%), cancer specific, and correlates with disease progression and outcome. CDO1 function can alternatively be silenced by repressive chromatin, and we describe protein-damaging missense mutations in 7% of tumors without DNA methylation. Restoration of CDO1 function in breast cancer cells increases levels of ROS and leads to reduced viability and growth, as well as sensitization to anthracycline treatment. Priming with 5-azacytidine of breast cancer cells with epigenetically silenced CDO1 resulted in restored expression and increased sensitivity to anthracyclines. CONCLUSION: We report that silencing of CDO1 is a critical epigenetic event that contributes to the survival of oxidative-stressed breast cancer cells through increased detoxification of ROS and thus leads to the resistance to ROS-generating chemotherapeutics including anthracyclines. Our study shows the importance of CDO1 inactivation in breast cancer and its clinical potential as a biomarker and therapeutic target to overcome resistance to anthracyclines.


Subject(s)
Anthracyclines/administration & dosage , Breast Neoplasms/genetics , Cysteine Dioxygenase/genetics , Drug Resistance, Neoplasm/immunology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cysteine Dioxygenase/antagonists & inhibitors , DNA Methylation/genetics , Drug Resistance, Neoplasm/genetics , Female , Gene Silencing , Humans , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...