Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Arch Biochem Biophys ; 591: 35-42, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26743849

ABSTRACT

Yeast alcohol dehydrogenase I is a homotetramer of subunits with 347 amino acid residues, catalyzing the oxidation of alcohols using NAD(+) as coenzyme. A new X-ray structure was determined at 3.0 Å where both subunits of an asymmetric dimer bind coenzyme and trifluoroethanol. The tetramer is a pair of back-to-back dimers. Subunit A has a closed conformation and can represent a Michaelis complex with an appropriate geometry for hydride transfer between coenzyme and alcohol, with the oxygen of 2,2,2-trifluoroethanol ligated at 2.1 Å to the catalytic zinc in the classical tetrahedral coordination with Cys-43, Cys-153, and His-66. Subunit B has an open conformation, and the coenzyme interacts with amino acid residues from the coenzyme binding domain, but not with residues from the catalytic domain. Coenzyme appears to bind to and dissociate from the open conformation. The catalytic zinc in subunit B has an alternative, inverted coordination with Cys-43, Cys-153, His-66 and the carboxylate of Glu-67, while the oxygen of trifluoroethanol is 3.5 Å from the zinc. Subunit B may represent an intermediate in the mechanism after coenzyme and alcohol bind and before the conformation changes to the closed form and the alcohol oxygen binds to the zinc and displaces Glu-67.


Subject(s)
Alcohol Dehydrogenase/chemistry , Alcohol Dehydrogenase/ultrastructure , NAD/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/ultrastructure , Trifluoroethanol/chemistry , Binding Sites , Catalysis , Coenzymes/chemistry , Coenzymes/ultrastructure , Computer Simulation , Enzyme Activation , Models, Chemical , Models, Molecular , NAD/ultrastructure , Protein Binding , Protein Conformation , Substrate Specificity
2.
Bioorg Med Chem ; 18(1): 134-41, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19959367

ABSTRACT

A series of indazole-dione derivatives were synthesized by the 1,3-dipolar cycloaddition reaction of appropriate substituted benzoquinones or naphthoquinones and N-carboalkoxyamino diazopropane derivatives. These compounds were evaluated for their effects on human carbonyl reductase. Several of the analogs were found to serve as substrates for carbonyl reductase with a wide range of catalytic efficiencies, while four analogs display inhibitory activities with IC(50) values ranging from 3-5 microM. Two of the inhibitors were studied in greater detail and were found to be noncompetitive inhibitors against both NADPH and menadione with K(I) values ranging between 2 and 11 microM. Computational studies suggest that conformation of the compounds may determine whether the indazole-diones bind productively to yield product or nonproductively to inhibit the enzyme.


Subject(s)
Alcohol Oxidoreductases/antagonists & inhibitors , Alcohol Oxidoreductases/metabolism , Indazoles/chemistry , Indazoles/pharmacology , Liver/enzymology , Humans , Inhibitory Concentration 50 , Structure-Activity Relationship
3.
Cardiovasc Toxicol ; 5(4): 365-76, 2005.
Article in English | MEDLINE | ID: mdl-16382174

ABSTRACT

Carbonyl reductase (CR) catalyzes the nicotinamide adenine dinucleotide phosphate (NADPH)-dependent reduction of several carbonyls. Anthracyclines used to treat cancer are reduced by CR at the C13 carbonyl and the resulting metabolites are implicated in the cardiotoxicity associated with anthracycline therapy. CR also is believed to have a role in detoxifying quinones, raising the question whether CR catalyzes reduction of anthracycline quinones. Steady-state kinetic studies were done with several anthraquinone-containing compounds, including 13-deoxydoxorubicin and daunorubicinol, which lack the C13 carbonyl, thus unmasking the anthraquinone for study. k(cat) and k(cat)/K(m) values for 13-deoxydoxorubicin and daunorubicinol were nearly identical, indicating that that the efficiency of quinone reduction was unaffected by the differences at the C13 position. k(cat) and k(cat)/K(m) values were much smaller for the analogs than for the parent compounds, suggesting that the C13 carbonyl is preferred as a substrate over the quinone. CR also reduced structurally related quinone molecules with less favorable catalytic efficiency. Modeling studies with doxorubicin and carbonyl reductase revealed that methionine 234 sterically hinder the rings adjacent to the quinone, thus accounting for the lower catalytic efficiency. Reduction of the anthraquinones may further define the role of CR in anthracycline metabolism and may influence anthracycline cytotoxic and cardiotoxic mechanisms.


Subject(s)
Alcohol Oxidoreductases/metabolism , Daunorubicin/analogs & derivatives , Doxorubicin/analogs & derivatives , Alcohol Oxidoreductases/isolation & purification , Animals , Anthraquinones/chemistry , Daunorubicin/chemistry , Daunorubicin/metabolism , Doxorubicin/metabolism , Humans , Models, Molecular , Oxidation-Reduction , Protein Structure, Tertiary , Recombinant Proteins
4.
Cardiovasc Toxicol ; 5(3): 269-83, 2005.
Article in English | MEDLINE | ID: mdl-16244372

ABSTRACT

Utilizing a model of chronic doxorubicin cardiomyopathy, this study examines the relationship between changes in expression and function of calcium handling proteins and contractile dysfunction. A possible mechanism to account for this relationship is suggested. New Zealand white rabbits were injected with either doxorubicin (1 mg/kg, twice weekly for 8 wk) or 0.9% NaCl. Gene transcript, protein levels, and the function of several proteins from the left ventricle were assessed. Protein levels of sarcoplasmic reticulum (SR) Ca2+ transporting ATPase (SERCA2a and b), Ca2+ release channel (RYR2), calsequestrin, Na/Ca exchanger, mRNA levels of RYR2, and [3H]-ryanodine binding (B(max)) to RYR2 were significantly decreased in doxorubicin-treated rabbits; protein levels of phospholamban, dihydropyridine receptor alpha2 subunit, and SR Ca2+ loading rates were not decreased. However, only protein levels of SERCA2 and RYR2, mRNA levels of RYR2, and Bmax of RYR2 significantly regressed with left-ventricular fractional shortening. Analysis of contractile function of atrial preparations isolated from doxorubicin-treated rabbits revealed that doxorubicin diminished contractility (dF/dt) of rest-potentiated contractions consistent with SR dysfunction. Serum concentrations of free triiodothyronine (T3) decreased in doxorubicin-treated rabbits. Our results suggest that chronic doxorubicin administration in the rabbit causes a SR-dependent contractile dysfunction that may result, in part, from decreased T3.


Subject(s)
Antibiotics, Antineoplastic/toxicity , Calcium/metabolism , Doxorubicin/toxicity , Heart Diseases/chemically induced , Sarcoplasmic Reticulum/drug effects , Triiodothyronine/metabolism , Animals , Antibodies, Monoclonal , Blotting, Western , Calcium Channels, L-Type/metabolism , Calcium-Binding Proteins/metabolism , Calcium-Transporting ATPases/metabolism , Calsequestrin/metabolism , Electrocardiography , Heart Diseases/physiopathology , Male , Myocardial Contraction/drug effects , Nuclease Protection Assays , Proteins/metabolism , RNA/biosynthesis , RNA/genetics , Rabbits , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcolemma/drug effects , Sarcolemma/metabolism , Sarcoplasmic Reticulum/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases , Sodium-Calcium Exchanger/metabolism , Thyrotropin/blood , Triiodothyronine/blood
5.
Mol Pharmacol ; 67(5): 1505-12, 2005 May.
Article in English | MEDLINE | ID: mdl-15705743

ABSTRACT

Anthracycline therapy is associated with a life-threatening but poorly understood cardiotoxicity. Effects of treatment are consistent with drug-induced disruption of cardiac sarcoplasmic reticulum (SR) calcium homeostasis, including inhibition of calcium release by anthracyclines. This effect, which depends on luminal SR calcium concentration, is hypothesized to involve interactions of anthracyclines with the calcium binding protein calsequestrin (CSQ). This study was designed to test the hypothesis that an interaction between CSQ and anthracyclines could be related to alterations in SR calcium release and cardiac function. The effects of the anthracycline, daunorubicin, and its metabolite daunorubicinol were compared with those of a known CSQ inhibitor, trifluoperazine (TFP). Protein fluorescence quenching studies demonstrated that TFP, daunorubicin, and daunorubicinol bind to CSQ with apparent binding affinities in the low micromolar range. The presence of calcium decreases the drug-dependent fluorescence quenching, probably because of calcium-induced CSQ conformational changes. TFP also inhibited SR calcium release. Although the TFP IC50 value is somewhat larger than for anthracyclines, the TFP effect is also dependent on luminal SR calcium concentration. In a muscle preparation, micromolar TFP decreased cardiac contractility in a manner that implicates the involvement of SR calcium and resembles the effects of anthracyclines. These data are consistent with a mechanism in which TFP or anthracyclines impair SR calcium release and cardiac function through a mechanism involving disruption of CSQ function. Such a mechanism may contribute to anthracycline cardiotoxicity.


Subject(s)
Calsequestrin/metabolism , Daunorubicin/analogs & derivatives , Daunorubicin/administration & dosage , Drug Delivery Systems/methods , Trifluoperazine/administration & dosage , Animals , Anthracyclines/administration & dosage , Anthracyclines/chemistry , Anthracyclines/metabolism , Daunorubicin/chemistry , Daunorubicin/metabolism , Dogs , Dose-Response Relationship, Drug , Myocardial Contraction/drug effects , Myocardial Contraction/physiology , Rabbits , Trifluoperazine/chemistry , Trifluoperazine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...