Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Singapore Med J ; 51(3): e58-61, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20428735

ABSTRACT

Urine pregnancy tests are widely used in emergency departments as the first screening test for patients of reproductive age presenting with gynaecological problems in order to rule out pregnancy. Conditions such as complete molar pregnancy, which produces large amounts of beta human chorionic gonadotropin (beta-hCG), may cause a false negative result due to an oversaturation of the assay system, known as the "hook effect". We report a case where the exclusion of pregnancy by urine testing led to the initial misdiagnosis of a molar pregnancy as a degenerative fibroid. Physicians need to be reminded of the possibility of false negative results with this commonly used test. Negative or inconclusive results in patients with a high suspicion of pregnancy should be further evaluated by serum quantification of beta-hCG and appropriate sample dilution.


Subject(s)
Chorionic Gonadotropin/urine , Hydatidiform Mole/diagnosis , Uterine Neoplasms/diagnosis , Biomarkers, Tumor , Chorionic Gonadotropin/blood , Chorionic Gonadotropin, beta Subunit, Human , Diagnostic Errors , False Negative Reactions , Female , Humans , Hydatidiform Mole/surgery , Leiomyoma/diagnosis , Middle Aged , Peptide Fragments , Pregnancy , Pregnancy Tests , Uterine Neoplasms/surgery
2.
Curr Alzheimer Res ; 4(4): 386-96, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17908041

ABSTRACT

Alzheimer's disease (AD) is linked to cholinergic deficiency and the overactivation of glutamate receptors. The acetylcholinesterase (AChE) inhibition treatment approach has produced the most encouraging results in clinical practice, and memantine, a moderate antagonist of N-methyl-D-aspartate (NMDA) receptors, has been approved for treating AD. However, AChE inhibitors have limited success as they only improve memory in mild dementia but cannot stop the process of neurodegeneration; while memantine possesses neuroprotective effects only with a little ability in memory enhancement. There has been a major rush among neuroscience research institutions and pharmaceutical firms worldwide to search for safer and more effective therapeutic agents for AD. The novel dimers, derived from tacrine and the fragment of huperzine A (HA'), have been demonstrated to be potent and selective reversible inhibitors of AChE. Bis(7)-tacrine, bis(12)-hupyridone (E12E) and HA'(10)-tacrine, are representatives of three series of novel dimers. According to the preclinical studies, these compounds have been shown to have low toxicity and high efficacy for improving cognitive deficits in several animal models. More interestingly, bis(7)-tacrine, similar to memantine, prevents glutamate-induced neurotoxicity by moderately blocking glutamate receptor NMDA subtype. Furthermore, bis(7)-tacrine, as well as E12E, possesses multiple neuroprotective effects in vitro and in vivo. Taking together, these dimeric AChE inhibitors, especially bis(7)-tacrine, E12E and HA'(10)-tacrine, may provide beneficial effects in AD and other neurodegenerative diseases.


Subject(s)
Alzheimer Disease/drug therapy , Cholinesterase Inhibitors/therapeutic use , Sesquiterpenes/therapeutic use , Tacrine/therapeutic use , Alkaloids , Animals , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/therapeutic use , Humans , Models, Biological , Sesquiterpenes/chemical synthesis , Sesquiterpenes/chemistry , Tacrine/chemical synthesis , Tacrine/chemistry
3.
Clin Pharmacol Ther ; 82(3): 282-93, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17443135

ABSTRACT

Organophosphorus compound-based nerve agents inhibit the essential enzyme acetylcholinesterase (AChE) causing acute toxicity and death. Clinical treatment of nerve-agent poisoning is to use oxime-based antidotes to reactivate the inhibited AChE. However, the nerve agent tabun is resistant to oximes. To design improved oximes, crystal structures of a tabun-conjugated AChE in complex with different oximes are needed to guide the structural modifications of known antidotes. However, this type of structure is extremely challenging to obtain because both deamidation of the tabun conjugate and reactivation of AChE occur during crystallographic experiments. Here we report, for the first time, the crystal structures of Ortho-7 and HLö-7 in complex with AChE that is conjugated to an intact tabun. These structures were determined by our new strategy of combining crystallographic and mass spectrometric analyses of AChE crystals. The results explain the relative reactivation potencies of the two oximes and offer insights into improving known medical antidotes.


Subject(s)
Acetylcholinesterase/chemistry , Antidotes/chemistry , Antidotes/pharmacology , Chemical Warfare Agents , Cholinesterase Inhibitors/chemistry , Organophosphates/antagonists & inhibitors , Organophosphates/chemistry , Chromatography, High Pressure Liquid , Crystallography, X-Ray , Drug Design , Mass Spectrometry , Models, Molecular , Molecular Conformation , Oximes/chemistry , Pyridines/chemistry , Pyridines/toxicity , Pyridinium Compounds/chemistry , Pyridinium Compounds/toxicity
4.
Clin Pharmacol Ther ; 81(1): 30-4, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17185996

ABSTRACT

Advances in genomics, proteomics, and structural genomics have identified a large number of protein targets. Virtual screening has gained popularity in identifying drug leads by computationally screening large numbers of chemicals against experimentally determined protein targets. In that context, there continues to be a "target-rich and lead-poor" imbalance, reflecting an insufficiency of chemists pursuing drug discovery in academia, the challenge of engaging more chemists in this area of research, and a paucity of available protein target structures. This imbalance in manpower and structural information can be ameliorated, in part, by adapting a "genome-to-drug-lead" approach, in which chemicals can be virtually screened against computer-predicted protein targets, within the context of the US National Science Foundation's petascale computing initiative. This approach offers a solution to reduce manpower requirements for more chemists to experimentally search for drug leads, which represent one of the greatest limitations to drug discovery and better exploits the extensive availability of drug targets at the gene level, ultimately improving the success of moving discoveries from the laboratory to the patient.


Subject(s)
Chemistry, Pharmaceutical/organization & administration , Drug Design , Genome, Human , Pharmacogenetics , Chemistry, Pharmaceutical/instrumentation , Computational Biology , Humans , Models, Molecular , Protein Conformation , Proteins/chemistry
5.
J Pharmacol Exp Ther ; 307(1): 190-6, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12893843

ABSTRACT

Recently, several bis-pyridiniumaldoximes linked by a variable-length alkylene chain were rationally designed in our laboratories as cholinesterase reactivators. Extensive in vitro tests of these oximes with acetylcholinesterase inhibited by two different organophosphate agents, echothiophate and diisopropylfluorophosphate, revealed one compound with particularly good reactivation kinetics and affinity for phosphorylated acetylcholinesterase (AChE). This compound, designated "ortho-7", with a heptylene chain bridging two aldoximes ortho to a pyridinium ring nitrogen, was chosen for detailed comparison with the classic reactivator pyridine-2-aldoxime methochloride (2-PAM). In vitro, ortho-7 reactivated AChE selectively, without restoring activity of the related enzyme butyrylcholinesterase (BChE). For in vivo studies, rats were injected with ortho-7 or 2-PAM before or after organophosphate exposure, and the activities of AChE and BChE were determined at multiple intervals in blood and solid tissues. Ortho-7 behaved nearly as well in the animal as in vitro, reactivating AChE to the same extent as 2-PAM in all peripheral tissues studied (serum, red blood cell, and diaphragm), but at doses up to 100-fold smaller. Like other oxime reactivators, ortho-7 did not reactivate brain AChE after systemic administration. Nonetheless, this agent could be useful in combination therapy for organophosphate exposure, and it may provide a platform for development of additional, even more effective reactivators.


Subject(s)
Cholinesterases/metabolism , Drug Design , Enzyme Reactivators/pharmacology , Oximes/pharmacology , Acetylcholinesterase/metabolism , Animals , Antidotes , Butyrylcholinesterase/metabolism , Computer-Aided Design , Enzyme Activation/drug effects , Enzyme Reactivators/toxicity , Kinetics , Male , Organophosphorus Compounds/antagonists & inhibitors , Organophosphorus Compounds/toxicity , Oximes/toxicity , Rats , Rats, Sprague-Dawley
6.
Planta Med ; 68(3): 217-20, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11914957

ABSTRACT

Intragastric administration (100-200 micromol/kg) of tacrine (THA) or bis(7)-THA could cause an acute and dose-dependent increase in plasma alanine aminotransferases activity in mice at 6 h after the drug administration. The increase in plasma enzyme activity was associated with an increase in hepatic malondialdehyde level, an indirect index of oxidative tissue damage. Pretreating mice with schisandrin B (Sch B), an active dibenzocyclooctadiene derivative isolated from the fruit of Schisandra chinensis, at a daily dose of 0.125-0.5 mmol/kg for 3 days protected against the THA/bis(7)-THA induced hepatic oxidative damage in a dose-dependent manner. Sch B treatment (0.025-0.5 mmol/kg/day x 5) also enhanced the passive avoidance-response in mice as assessed by the step-through task experiment. The ensemble of results suggests that Sch B may be useful for reducing the potential hepatotoxicity of THA/bis(7)-THA in anti-Alzheimer's therapy.


Subject(s)
Cognition/drug effects , Lignans/pharmacology , Liver/drug effects , Polycyclic Compounds/pharmacology , Schisandraceae , Tacrine/analogs & derivatives , Tacrine/toxicity , Alanine Transaminase/blood , Alanine Transaminase/metabolism , Animals , Avoidance Learning/drug effects , Behavior, Animal/drug effects , Cyclooctanes , Dose-Response Relationship, Drug , Fruit/chemistry , Lignans/chemistry , Lignans/isolation & purification , Male , Mice , Mice, Inbred ICR , Plant Extracts/chemistry , Plant Extracts/pharmacology , Polycyclic Compounds/chemistry , Polycyclic Compounds/isolation & purification , alpha-Tocopherol/pharmacology
7.
FEBS Lett ; 502(3): 93-7, 2001 Aug 03.
Article in English | MEDLINE | ID: mdl-11583118

ABSTRACT

Using the computer docking program EUDOC, in silico screening of a chemical database for inhibitors of human adenovirus cysteine proteinase (hAVCP) identified 2,4,5,7-tetranitro-9-fluorenone that selectively and irreversibly inhibits hAVCP in a two-step reaction: reversible binding (Ki = 3.09 microM) followed by irreversible inhibition (ki = 0.006 s(-1)). The reversible binding is due to molecular complementarity between the inhibitor and the active site of hAVCP, which confers the selectivity of the inhibitor. The irreversible inhibition is due to substitution of a nitro group of the inhibitor by the nearby Cys122 in the active site of hAVCP. These findings suggest a new approach to selective, irreversible inhibitors of cysteine proteinases involved in normal and abnormal physiological processes ranging from embryogenesis to apoptosis and pathogen invasions.


Subject(s)
Adenoviruses, Human/enzymology , Cysteine Endopeptidases/drug effects , Cysteine Proteinase Inhibitors/pharmacology , Fluorenes/pharmacology , Animals , Cattle , Cysteine/chemistry , Cysteine Endopeptidases/metabolism , Cysteine Proteinase Inhibitors/chemical synthesis , Drug Design , Drug Evaluation, Preclinical , Fluorenes/chemical synthesis , Humans , Molecular Structure , Papain/drug effects , Recombinant Proteins/isolation & purification
8.
Proteins ; 45(3): 183-9, 2001 Nov 15.
Article in English | MEDLINE | ID: mdl-11599021

ABSTRACT

I report herein two 2.0 ns (1.0 fs time step) MD simulations of two zinc complexes bridged by a hydroxide in phosphotriesterase (PTE) employing the nonbonded method and the cationic dummy atom method that uses virtual atoms to impose orientational requirement for zinc ligands. The cationic dummy atom method was able to simulate the four-ligand coordination of the two zinc complexes in PTE. The distance (3.39 +/- 0.07A) between two nearby zinc ions in the time-average structure of PTE derived from the MD simulation using the cationic dummy atoms matched that in the X-ray structure (3.31 +/- 0.001A). Unequivocally, the time-average structure of PTE was able to fit into the experimentally determined difference electron density map of the corresponding X-ray structure. The results demonstrate the practicality of the cationic dummy atom method for MD simulations of zinc proteins bound with multiple zinc ions. In contrast, a 2.0 ns (1.0 fs time step) MD simulation using the nonbonded method revealed a striking difference in the active site between the X-ray structure and the time-average structure that was unable to fit into the density map of PTE. The results suggest that caution should be used in the MD simulations using the nonbonded method.


Subject(s)
Cations, Divalent/chemistry , Esterases/chemistry , Hydroxides/chemistry , Zinc/chemistry , Algorithms , Aryldialkylphosphatase , Computer Simulation , Crystallography, X-Ray/methods , Models, Molecular , Organophosphonates/chemistry , Protein Conformation , Thermodynamics
9.
Biochem Biophys Res Commun ; 287(1): 204-8, 2001 Sep 14.
Article in English | MEDLINE | ID: mdl-11549275

ABSTRACT

Histamine N-methyltransferase (HNMT) catalyzes the N-methylation of histamine in mammals. The experimentally determined HNMT three-dimensional (3D) structure is not available. However, there is a common genetic polymorphism for human HNMT (Thr105Ile) that reduces enzymatic activity and is a risk factor for asthma. To obtain insights into mechanisms responsible for the effects of that polymorphism on enzymatic activity and thermal stability, we predicted the 3D structure of HNMT using the threading method and molecular dynamics simulations in water. Herein, we report a theoretical 3D model of human HNMT which reveals that polymorphic residue Thr105Ile is located in the turn between a beta strand and an alpha helix on the protein surface away from the active site of HNMT. Ile105 energetically destabilizes folded HNMT because of its low Chou-Fasman score for forming a turn conformation and the exposure of its hydrophobic side chain to aqueous solution. It thus promotes the formation of misfolded proteins that are prone to the clearance by proteasomes. This information explains, for the first time, how genetic polymorphisms can cause enhanced protein degradation and why the thermal stability of allozyme Ile105 is lower than that of Thr105. It also supports the hypothesis that the experimental observation of a significantly lower level of HNMT enzymatic activity for allozyme Ile105 than that with Thr105 is due to a decreased concentration of allozyme Ile105, but not an alternation of the active-site topology of HNMT caused by the difference at residue 105.


Subject(s)
Histamine N-Methyltransferase/metabolism , Models, Theoretical , Computer Simulation , Enzyme Stability , Gene Deletion , Histamine N-Methyltransferase/chemistry , Histamine N-Methyltransferase/genetics , Humans , Models, Molecular , Polymorphism, Single Nucleotide , Protein Conformation , Temperature
10.
Mol Vis ; 7: 164-71, 2001 Jul 18.
Article in English | MEDLINE | ID: mdl-11483892

ABSTRACT

PURPOSE: To synthesize novel aldose reductase inhibitors (ARI) that will normalize losses in protein kinase Cgamma (PKCgamma) observed during diabetes and galactosemia. METHODS: ARI were synthesized as tricyclic pyrones 1-6 (HAR-1 through HAR-6) from 3-methyl-1H,7H-5a,6,8,9-tetrahydro-1-oxopyrano[4,3-b][1]benzopyran and (5aS,7S)-7-isopropenyl-3-methyl-1H,7H-5a,6,8,9-tetrahydro-1-oxopyrano[4,3-b][1]benzopyran and were tested by inhibition of aldose reductase enzyme activity in vitro and by inhibition of polyol formation in lens epithelial cells in culture. Identified compounds were further tested in galactosemic rat lens in vivo for (a) normalized PKCgamma levels by Western blot, (b) reduction of phosphorylation of the gap junction protein Cx46 by analyses of co-immunoprecipitated proteins, and (c) by normalization of gap junction activity as measured by dye transfer. RESULTS: HAR-1 (1H,7H-5a,6,8,9-tetrahydro-1-oxopyrano[4,3-b][1]benzopyran-3-acetic acid) was identified as an ARI with IC50 for aldose reductase inhibition at 2 nM. Polyol accumulation in lens epithelial cells was reduced by 80% at 10 microM. Rats fed 40% galactose for 9 days had an 80% reduction in PKCgamma levels which were normalized by HAR-1 at 100 mg/kg/day, fed orally. Phosphorylation of Cx46 was increased by 50% and this was normalized in HAR-1 treated rats (6 day treatment). Gap junction activity of galactosemic rats was reduced by 55% and this was normalized by HAR-1 in six day-treated rats. CONCLUSIONS: HAR-1 is a novel ARI which normalized losses of PKCgamma, changes in Cx46 phosphorylation, and gap junction activity.


Subject(s)
Acetates/pharmacology , Aldehyde Reductase/antagonists & inhibitors , Benzopyrans/pharmacology , Enzyme Inhibitors/pharmacology , Isoenzymes/metabolism , Lens, Crystalline/drug effects , Protein Kinase C/metabolism , Acetates/chemical synthesis , Animals , Benzopyrans/chemical synthesis , Blotting, Western , Connexins/metabolism , Diabetes Mellitus/enzymology , Drug Evaluation, Preclinical , Enzyme Inhibitors/chemical synthesis , Epithelial Cells/metabolism , Galactose/administration & dosage , Galactosemias/enzymology , Gap Junctions/metabolism , Lens Cortex, Crystalline/metabolism , Lens, Crystalline/enzymology , Molecular Structure , Phosphorylation , Polymers/metabolism , Rabbits , Rats , Rats, Sprague-Dawley , Serine/metabolism
11.
J Neurophysiol ; 86(1): 183-9, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11431500

ABSTRACT

Bis(7)-tacrine is a potent acetylcholinesterase inhibitor in which two tacrine molecules are linked by a heptylene chain. We tested the effects of bis(7)-tacrine on the spontaneous synaptic activity. Miniature endplate potentials (MEPPs) were recorded extracellularly on slices of electric organ of Torpedo marmorata. Bis(7)-tacrine, at a concentration of 100 nM, increased the magnitudes that describe MEPPs: amplitude, area, rise time, rate of rise, and half-width. We also tested the effect of bis(7)-tacrine on nicotinic acetylcholine receptors by analyzing the currents elicited by acetylcholine (100 microM) in Torpedo electric organ membranes transplanted in Xenopus laevis oocytes. Bis(7)-tacrine inhibited the acetylcholine-induced currents in a reversible manner (IC(50) = 162 nM). The inhibition of nicotinic acetylcholine receptors was not voltage dependent, and bis(7)-tacrine increased the desensitization of nicotinic acetylcholine receptors. The Hill coefficient for bis(7)-tacrine was -0.72 +/- 0.02, indicating that bis(7)-tacrine binds to the nicotinic acetylcholine receptor in a molecular ratio of 1:1, but does not affect the binding of alpha-bungarotoxin with the nicotinic acetylcholine receptor. In conclusion, bis(7)-tacrine greatly increases the spontaneous quantal release from peripheral cholinergic terminals at a much lower concentration than tacrine. Bis(7)-tacrine also blocks acetylcholine-induced currents of Torpedo electric organ, although the mechanism is different from that of tacrine: bis(7)-tacrine enhances desensitization, whereas tacrine reduces it.


Subject(s)
Cholinesterase Inhibitors/pharmacology , Electric Organ/physiology , Receptors, Nicotinic/drug effects , Synaptic Transmission/drug effects , Tacrine/analogs & derivatives , Tacrine/pharmacology , Animals , Bungarotoxins/metabolism , Bungarotoxins/pharmacology , Cholinesterase Inhibitors/chemistry , Dose-Response Relationship, Drug , Electric Organ/drug effects , Female , Iodine Radioisotopes , Membrane Potentials/drug effects , Membrane Potentials/physiology , Membrane Proteins/metabolism , Oocytes/physiology , Oocytes/transplantation , Receptors, Nicotinic/metabolism , Solubility , Tacrine/chemistry , Torpedo , Xenopus laevis
12.
Curr Med Chem ; 8(8): 933-9, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11375760

ABSTRACT

Combination therapy in the treatment of viral infections in which, for example, three different drugs against three different targets on three independent proteins are administered, has been highly successful clinically. However, it is only a matter of time before a virus will arise resistant to all three drugs, because the mutations leading to drug resistance are independent of each other. But, what if the mutations leading to drug resistance are not independent of each other, but confer some cost to the virus? If the cost is too great, than resistance may not arise. To impose such a cost in the clinical treatment of viral infections, we propose a new form of combination therapy. Here, three different drugs against three different targets on the same virus-coded protein are administered. If the physiological functions of the three different target sites are not independent of each other, then, a mutation at one site may alter the physiological functions at the other sites. We present a model system in which to test the efficacy of this new form of triple combination therapy. Human adenovirus has a virus-coded proteinase that is essential for the synthesis of infectious virus. It contains an active site and two cofactor binding sites; the functions of the active site are dependent upon the cofactors interacting with their binding sites. We describe how to obtain drugs against the three different sites.


Subject(s)
Anti-HIV Agents/therapeutic use , HIV Infections/drug therapy , Models, Biological , Adenoviridae/enzymology , Binding Sites , Cysteine Endopeptidases/chemistry , Cysteine Endopeptidases/drug effects , Cysteine Endopeptidases/genetics , DNA/drug effects , DNA/metabolism , Drug Resistance, Microbial/physiology , Drug Therapy, Combination , HIV/drug effects , HIV/enzymology , HIV Integrase Inhibitors/therapeutic use , HIV Protease Inhibitors/therapeutic use , Humans , Reverse Transcriptase Inhibitors/therapeutic use
13.
J Biol Chem ; 276(12): 9330-6, 2001 Mar 23.
Article in English | MEDLINE | ID: mdl-11104759

ABSTRACT

Butyrylcholinesterase (BChE) is important in cocaine metabolism, but it hydrolyzes (-)-cocaine only one-two thousandth as fast as the unnatural (+)-stereoisomer. A starting point in engineering BChE mutants that rapidly clear cocaine from the bloodstream, for overdose treatment, is to elucidate structural factors underlying the stereochemical difference in catalysis. Here, we report two three-dimensional Michaelis-Menten complexes of BChE liganded with natural and unnatural cocaine molecules, respectively, that were derived from molecular modeling and supported by experimental studies. Such complexes revealed that the benzoic ester group of both cocaine stereoisomers must rotate toward the catalytic Ser(198) for hydrolysis. Rotation of (-)-cocaine appears to be hindered by interactions of its phenyl ring with Phe(329) and Trp(430). These interactions do not occur with (+)-cocaine. Because the rate of (-)-cocaine hydrolysis is predicted to be determined mainly by the re-orientation step, it should not be greatly influenced by pH. In fact, measured rates of this reaction were nearly constant over the pH range from 5.5 to 8.5, despite large rate changes in hydrolysis of (+)-cocaine. Our models can explain why BChE hydrolyzes (+)-cocaine faster than (-)-cocaine, and they suggest that mutations of certain residues in the catalytic site could greatly improve catalytic efficiency and the potential for detoxication.


Subject(s)
Butyrylcholinesterase/metabolism , Cocaine/pharmacokinetics , Mutation , Butyrylcholinesterase/chemistry , Butyrylcholinesterase/genetics , Cocaine/chemistry , Hydrogen-Ion Concentration , Hydrolysis , Inactivation, Metabolic , Models, Molecular , Molecular Conformation , Protein Conformation
14.
Protein Sci ; 9(10): 1857-65, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11106157

ABSTRACT

Farnesyltransferase (FT) inhibitors can suppress tumor cell proliferation without substantially interfering with normal cell growth, thus holding promise for cancer treatment. A structure-based approach to the design of improved FT inhibitors relies on knowledge of the conformational flexibility of the zinc-containing active site of FT. Although several X-ray structures of FT have been reported, detailed information regarding the active site conformational flexibility of the enzyme is still not available. Molecular dynamics (MD) simulations of FT can offer the requisite information, but have not been applied due to a lack of effective methods for simulating the four-ligand coordination of zinc in proteins. Here, we report in detail the problems that occurred in the conventional MD simulations of the zinc-bound FT and a solution to these problems by employing a simple method that uses cationic dummy atoms to impose orientational requirement for zinc ligands. A successful 1.0 ns (1.0 fs time step) MD simulation of zinc-bound FT suggests that nine conserved residues (Asn127alpha, Gln162alpha, Asn165alpha, Gln195alpha, His248beta, Lys294beta, Leu295beta, Lys353beta, and Ser357beta) in the active site of mammalian FT are relatively mobile. Some of these residues might be involved in the ligand-induced active site conformational rearrangement upon binding and deserve attention in screening and design of improved FT inhibitors for cancer chemotherapy.


Subject(s)
Alkyl and Aryl Transferases/chemistry , Alkyl and Aryl Transferases/metabolism , Computer Simulation , Enzyme Inhibitors/chemistry , Zinc/metabolism , Alkyl and Aryl Transferases/antagonists & inhibitors , Amino Acid Sequence , Animals , Binding Sites , Conserved Sequence , Crystallography, X-Ray , Drug Design , Farnesyltranstransferase , Models, Molecular , Protein Conformation , Rats , Zinc/chemistry
15.
Neurosci Lett ; 288(2): 95-8, 2000 Jul 14.
Article in English | MEDLINE | ID: mdl-10876069

ABSTRACT

The effects of bis(7)-tacrine, a novel acetylcholinesterase inhibitor, on ischemia-induced cell death and apoptosis were investigated in primary cerebral cortical astrocytes of mice. Following a 6 h in vitro ischemic incubation of the cultures, a marked decrease in the percentage of viable cells was observed by lactate dehydrogenase (LDH) release assay. Furthermore, using bisbenzimide staining, we determined that approximately 65% of the cells underwent apoptosis. Treatment with bis(7)-tacrine (1-10 nM) during ischemic incubation effectively inhibited the ischemia-induced apoptosis, as demonstrated by morphological and biochemical tests. Our results demonstrated that bis(7)-tacrine could protect astrocytes against ischemia-induced cell injury, indicating that the drug might be beneficial for the treatment of vascular dementia, in addition to Alzheimer's disease.


Subject(s)
Alzheimer Disease/drug therapy , Astrocytes/pathology , Brain Ischemia/pathology , Brain Ischemia/prevention & control , Cerebral Cortex/pathology , Cholinesterase Inhibitors/therapeutic use , Tacrine/analogs & derivatives , Tacrine/therapeutic use , Animals , Animals, Newborn , Apoptosis/drug effects , Astrocytes/enzymology , Brain Ischemia/enzymology , Cells, Cultured , Cerebral Cortex/drug effects , Cerebral Cortex/enzymology , Female , Male , Mice , Mice, Inbred ICR
16.
Cancer Res ; 60(9): 2419-28, 2000 May 01.
Article in English | MEDLINE | ID: mdl-10811119

ABSTRACT

Flavopiridol, the first potent cyclin-dependent kinase inhibitor to enter clinical trials, was recently found to be cytotoxic to noncycling cells. The present studies were performed to examine the hypothesis that flavopiridol, like several other antineoplastic agents that kill noncycling cells, might also interact with DNA. Consistent with this possibility, treatment of A549 human lung cancer cells with clinically achievable concentrations of flavopiridol resulted in rapid elevations of the DNA damage-responsive protein p53. In further studies, the binding of flavopiridol to DNA was examined in vitro by four independent techniques. Absorption spectroscopy revealed that addition of DNA to aqueous flavopiridol solutions resulted in a red shift of the flavopiridol lambda(max) from 311 to 344 nm, demonstrating an isosbestic point typical of changes seen with DNA-binding compounds. Reverse-phase high-performance liquid chromatography demonstrated that flavopiridol binds to genomic DNA to a similar extent as ethidium bromide and Hoechst 33258. Nuclear magnetic resonance spectroscopy revealed that DNA caused extreme broadening of flavopiridol 1H nuclear magnetic resonance signals that could be reversed by addition of ethidium bromide or by DNA melting, suggesting that flavopiridol binds to (and likely intercalates into) duplex DNA. Equilibrium dialysis demonstrated that the equilibrium dissociation constant of the flavopiridol-DNA complex (5.4+/-3.4 x 10(-4) M) was in the same range observed for binding of the intercalators doxorubicin and pyrazoloacridine to DNA. Molecular modeling confirmed the feasibility of flavopiridol intercalation into DNA and analysis of the effects of flavopiridol in the National Cancer Institute tumor cell line panel using the COMPARE algorithm demonstrated that flavopiridol most closely resembles cytotoxic antineoplastic intercalators. Collectively, these data suggest that DNA might be a second target of flavopiridol, providing a potential explanation for the ability of this agent to kill noncycling cancer cells.


Subject(s)
Antineoplastic Agents/metabolism , DNA/metabolism , Enzyme Inhibitors/metabolism , Flavonoids/metabolism , Piperidines/metabolism , Acridines/pharmacology , Animals , Apigenin , Cattle , Chromatography, High Pressure Liquid , Cyclin-Dependent Kinases/antagonists & inhibitors , DNA/drug effects , DNA/radiation effects , DNA Topoisomerases, Type I/metabolism , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Enzyme Inhibitors/pharmacology , Ethidium/pharmacology , Flavonoids/pharmacology , Fluorescent Dyes/pharmacology , Humans , Immunoblotting , Intercalating Agents/pharmacology , Magnetic Resonance Spectroscopy , Models, Molecular , Piperidines/pharmacology , Pyrazoles/pharmacology , RNA/drug effects , RNA/metabolism , Time Factors , Tumor Cells, Cultured , Tumor Suppressor Protein p53/biosynthesis
17.
J Med Chem ; 43(3): 401-8, 2000 Feb 10.
Article in English | MEDLINE | ID: mdl-10669567

ABSTRACT

Virtual screening of chemical databases is an emerging approach in drug discovery that uses computers to dock chemicals into the active site of a drug target to identify leads through evaluation of binding affinities of the chemicals. However, there are concerns about the validity and scope of the reported virtual screens due to lack of studies to show that randomly selected chemicals are not equally active and due to the fact that metalloproteins were rarely used as drug targets. We have performed a virtual screening of a chemical database to identify prototypic inhibitors of farnesyltransferase (FT) with zinc present in the active site. Among the 21 compounds identified by computers, four inhibited FT in vitro with IC(50) values in the range from 25 to 100 microM. The most potent inhibitor also inhibited FT in human lung cancer cells. In contrast, none of 21 randomly selected compounds have an IC(50) lower than 100 microM. The results demonstrate the validity of virtual screening and the feasibility of applications of this approach to metalloprotein drug targets, such as matrix metalloproteinases, farnesyltransferase, and HIV-1 integrase, for the treatments of cardiovascular diseases, cancers, and AIDS.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Drug Design , Enzyme Inhibitors/chemistry , Alkyl and Aryl Transferases/metabolism , Animals , Binding Sites , Databases, Factual , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Farnesyltranstransferase , Humans , Models, Molecular , Rats , Tumor Cells, Cultured , Zinc/chemistry , Zinc/metabolism
18.
J Med Chem ; 42(20): 4225-31, 1999 Oct 07.
Article in English | MEDLINE | ID: mdl-10514292

ABSTRACT

Dimeric acetylcholinesterase (AChE) inhibitors containing a single 9-amino-1,2,3,4-tetrahydroacridine (tacrine) unit were constructed in an effort to further delineate structural requirements for optimal binding to the AChE peripheral site. Basic amines of differing hydrophobicity were selected as peripheral site ligands, and in each case, improvements in inhibitory potency and selectivity were seen relative to tacrine itself. AChE IC(50) values of the optimum dimers decrease significantly as the peripheral site ligand was permuted in the series ammonia > dimethylamine > 4-aminopyridine > 4-aminoquinoline > tacrine. Calculated desolvation free energies of the optimum dimers match the trend in IC(50) values, suggesting the importance of ligand hydrophobicity for effective cation-pi interaction with the peripheral site.


Subject(s)
Cholinesterase Inhibitors/chemical synthesis , Tacrine/analogs & derivatives , Tacrine/chemical synthesis , Acetylcholinesterase/metabolism , Animals , Butyrylcholinesterase/metabolism , Cholinesterase Inhibitors/chemistry , Cholinesterase Inhibitors/metabolism , Cholinesterase Inhibitors/pharmacology , Crystallography, X-Ray , Drug Design , In Vitro Techniques , Ligands , Protein Binding , Rats , Structure-Activity Relationship , Tacrine/chemistry , Tacrine/pharmacology
19.
Bioorg Med Chem Lett ; 9(16): 2335-8, 1999 Aug 16.
Article in English | MEDLINE | ID: mdl-10476864

ABSTRACT

Hybrid acetylcholinesterase inhibitors composed of a key fragment of huperzine A and an intact tacrine unit were prepared. The syntheses are quite direct, proceeding in a maximum of 4 linear steps from commercially available starting materials. The optimum hybrid inhibitor (+/-)-9g is 13-fold more potent than (-)-huperzine A, and 25-fold more potent than tacrine.


Subject(s)
Acetylcholinesterase/drug effects , Cholinesterase Inhibitors/pharmacology , Sesquiterpenes/chemistry , Tacrine/chemistry , Alkaloids , Cholinesterase Inhibitors/chemistry
20.
Zhongguo Yao Li Xue Bao ; 20(3): 211-7, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10452094

ABSTRACT

AIM: To study the effects of 1,7-N-heptylene-bis-9,9'-amino-1,2,3,4-tetrahydroacridine [bis(7)-tacrine], a novel dimeric acetylcholine-sterase inhibitor (AChEI) derived from 9-amino-1,2,3,4-tetrahydroaminoacridine (tacrine), on scopolamine-induced spatial memory impairment. METHODS: The effects of bis(7)-tacrine were investigated on the 5-d performance of young adult rats in the Morris water maze. The latency to find the platform in the water maze was measured to evaluate performance. Tacrine was used as a reference drug. RESULTS: Scopolamine (0.3 mg.kg-1, i.p.) resulted in an increase in latency period (> 100% increase) as compared with saline treated controls. Both bis(7)-tacrine and tacrine lessened the increased latency induced by scopolamine to the level of saline control group. The relative potency of bis(7)-tacrine (0.35 mumol.kg-1, i.g. or i.p.) to shorten the escape latency was 24 or 12 times of tacrine (8.52 mumol.kg-1 i.g., 4.26 mumol.kg-1 i.p.) following i.g. or i.p. administration, respectively. There appeared to be an inverse bell-shape dose-dependent effect for both compounds tested. CONCLUSION: Bis(7)-tacrine is a more potent and orally active AChEI than tacrine, and has potential for the palliative treatment of Alzheimer disease.


Subject(s)
Cholinesterase Inhibitors/pharmacology , Maze Learning/drug effects , Memory/drug effects , Tacrine/analogs & derivatives , Animals , Male , Rats , Rats, Sprague-Dawley , Scopolamine , Tacrine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...