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
2.
Nucleic Acids Res ; 42(Database issue): D1091-7, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24203711

ABSTRACT

DrugBank (http://www.drugbank.ca) is a comprehensive online database containing extensive biochemical and pharmacological information about drugs, their mechanisms and their targets. Since it was first described in 2006, DrugBank has rapidly evolved, both in response to user requests and in response to changing trends in drug research and development. Previous versions of DrugBank have been widely used to facilitate drug and in silico drug target discovery. The latest update, DrugBank 4.0, has been further expanded to contain data on drug metabolism, absorption, distribution, metabolism, excretion and toxicity (ADMET) and other kinds of quantitative structure activity relationships (QSAR) information. These enhancements are intended to facilitate research in xenobiotic metabolism (both prediction and characterization), pharmacokinetics, pharmacodynamics and drug design/discovery. For this release, >1200 drug metabolites (including their structures, names, activity, abundance and other detailed data) have been added along with >1300 drug metabolism reactions (including metabolizing enzymes and reaction types) and dozens of drug metabolism pathways. Another 30 predicted or measured ADMET parameters have been added to each DrugCard, bringing the average number of quantitative ADMET values for Food and Drug Administration-approved drugs close to 40. Referential nuclear magnetic resonance and MS spectra have been added for almost 400 drugs as well as spectral and mass matching tools to facilitate compound identification. This expanded collection of drug information is complemented by a number of new or improved search tools, including one that provides a simple analyses of drug-target, -enzyme and -transporter associations to provide insight on drug-drug interactions.


Subject(s)
Databases, Chemical , Drug Discovery , Pharmacokinetics , Internet , Pharmaceutical Preparations/chemistry , Quantitative Structure-Activity Relationship
3.
J Alzheimers Dis ; 28(2): 403-22, 2012.
Article in English | MEDLINE | ID: mdl-22045496

ABSTRACT

The concentration of presenilin-1 (PS-1) protein at the mitochondrial-associated aspect of the endoplasmic reticulum supports the potential for a mitochondrial influence of PS-1. Given that carriers of certain Alzheimer's disease (AD)-related PS-1 variants are predisposed to clinical depression and that depression has been historically associated with the mitochondrial enzyme, monoamine oxidase-A (MAO-A), we investigated cortical MAO-A function in the AD-related PS-1(M146V) knock-in mouse. The MAO-A system was clearly altered in the PS-1(M146V) mouse as revealed by (a) a mismatch between MAO-A protein expression and MAO-A activity; (b) changes in MAO-A-mediated monoaminergic neurotransmitter metabolism; (c) changes in non-cognitive behavior following treatment with the irreversible MAO-A inhibitor clorgyline; and (d) an increase in the potency of clorgyline in these same mice. We next investigated whether PS-1(M146V) could be influencing MAO-A directly. We observed (a) an enhanced MAO-A activity in necropsied PS-1(M146V) mouse cortical extracts incubated with DAPT (a PS-1 substrate-competitor); (b) the proximity of PS-1 with MAO-A and mitochondrial markers in cortical sections and in primary cortical neurons; (c) the co-segregation and co-immunoprecipitation of PS-1 and MAO-A within the mitochondrial fraction; and (d) the co-immunoprecipitation of overexpressed PS-1(M146V) and MAO-A proteins from N2a lysates. The PS-1(ΔEx9) and PS-1(D257A) variants, known to have low substrate-binding capacity, co-immunoprecipitated weakly with MAO-A. These combined data support a physical interaction between PS-1 and MAO-A that could influence MAO-A activity and contribute to the monoaminergic disruptions common to disorders as seemingly diverse as depression and AD.


Subject(s)
Alzheimer Disease/pathology , Cerebral Cortex/enzymology , Gene Expression Regulation, Enzymologic/genetics , Methionine/genetics , Monoamine Oxidase/metabolism , Presenilin-1/genetics , Valine/genetics , Alzheimer Disease/genetics , Analysis of Variance , Animals , Cells, Cultured , Cerebral Cortex/pathology , Chromatography, High Pressure Liquid/methods , Clorgyline/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Gene Expression Regulation, Enzymologic/drug effects , Hydroxyindoleacetic Acid/metabolism , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Mice , Mice, Transgenic , Mitochondria/metabolism , Mitochondria/ultrastructure , Monoamine Oxidase Inhibitors/pharmacology , Mutation/genetics , Neuroblastoma/pathology , Neurons/drug effects , Neurons/enzymology , Neurons/ultrastructure , Neurotransmitter Agents/metabolism , Organic Chemicals , Serotonin/metabolism , Subcellular Fractions/enzymology , Swimming/psychology , Transfection , Voltage-Dependent Anion Channels/metabolism
4.
J Neural Transm (Vienna) ; 118(7): 987-95, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21373759

ABSTRACT

Monoamine oxidase-A (MAO-A) has been associated with both depression and Alzheimer disease (AD). Recently, carriers of AD-related presenilin-1 (PS-1) alleles have been found to be at higher risk for developing clinical depression. We chose to examine whether PS-1 could influence MAO-A function in vitro. Overexpression of selected AD-related PS-1 variants (wildtype, Y115H, ΔEx9 and M146V) in mouse hippocampal HT-22 cells affects MAO-A catalytic activity in a variant-specific manner. The ability of the PS-1 substrate-competitor DAPT to induce MAO-A activity in cells expressing either PS-1 wildtype or PS-1(M146V) suggests the potential for a direct influence of PS-1 on MAO-A function. In support of this, we were able to co-immunoprecipitate MAO-A with FLAG-tagged PS-1 wildtype and M146V proteins. This potential for a direct protein-protein interaction between PS-1 and MAO-A is not specific for HT-22 cells as we were also able to co-immunoprecipitate MAO-A with FLAG-PS-1 variants in N2a mouse neuroblastoma cells and in HEK293 human embryonic kidney cells. Finally, we demonstrate that the two PS-1 variants reported to be associated with an increased incidence of clinical depression [e.g., A431E and L235V] both induce MAO-A activity in HT-22 cells. A direct influence of PS-1 variants on MAO-A function could provide an explanation for the changes in monoaminergic tone observed in several neurodegenerative processes including AD. The ability to induce MAO-A catalytic activity with a PS-1/γ-secretase inhibitor should also be considered when designing secretase inhibitor-based therapeutics.


Subject(s)
Alzheimer Disease/enzymology , Depressive Disorder/enzymology , Genetic Variation , Monoamine Oxidase/metabolism , Neurons/enzymology , Presenilin-1/genetics , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Animals , Cell Line, Transformed , Cell Line, Tumor , Depressive Disorder/genetics , Depressive Disorder/pathology , HEK293 Cells , Humans , Mice , Neuroblastoma/enzymology , Neuroblastoma/pathology , Neurons/cytology , Presenilin-1/physiology
5.
BMC Neurosci ; 8: 73, 2007 Sep 16.
Article in English | MEDLINE | ID: mdl-17868476

ABSTRACT

BACKGROUND: Calcium (Ca2+) has recently been shown to selectively increase the activity of monoamine oxidase-A (MAO-A), a mitochondria-bound enzyme that generates peroxyradicals as a natural by-product of the deamination of neurotransmitters such as serotonin. It has also been suggested that increased intracellular free Ca2+ levels as well as MAO-A may be contributing to the oxidative stress associated with Alzheimer disease (AD). RESULTS: Incubation with Ca2+ selectively increases MAO-A enzymatic activity in protein extracts from mouse hippocampal HT-22 cell cultures. Treatment of HT-22 cultures with the Ca2+ ionophore A23187 also increases MAO-A activity, whereas overexpression of calbindin-D28K (CB-28K), a Ca2+-binding protein in brain that is greatly reduced in AD, decreases MAO-A activity. The effects of A23187 and CB-28K are both independent of any change in MAO-A protein or gene expression. The toxicity (via production of peroxyradicals and/or chromatin condensation) associated with either A23187 or the AD-related beta-amyloid peptide, which also increases free intracellular Ca2+, is attenuated by MAO-A inhibition in HT-22 cells as well as in primary hippocampal cultures. CONCLUSION: These data suggest that increases in intracellular Ca2+ availability could contribute to a MAO-A-mediated mechanism with a role in AD-related oxidative stress.


Subject(s)
Alzheimer Disease/enzymology , Alzheimer Disease/pathology , Calcium/physiology , Free Radicals/metabolism , Hippocampus/enzymology , Hippocampus/pathology , Monoamine Oxidase/metabolism , Peroxides/metabolism , Alzheimer Disease/metabolism , Animals , Cell Line, Transformed , Cells, Cultured , Hippocampus/metabolism , Mice , Monoamine Oxidase/physiology , Oxidative Stress/physiology , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...