Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Inflammopharmacology ; 11(2): 183-8, 2003.
Article in English | MEDLINE | ID: mdl-15035820

ABSTRACT

Prompted by the findings that smokers have lowered brain and blood platelet monoamine oxidase-A and -B activities compared to non-smokers and that smokers have a lowered incidence of Parkinson's disease, we have examined the neuroprotective properties of an MAO inhibitor, 2,3,6-trimethyl-1,4-naphthoquinone (TMN), which is present in the tobacco plant and smoke in the MPTP C57BL/6 mouse model of neurodegeneration. Dopamine (DA) levels in the striata of mice treated with TMN prior to the administration of MPTP were significantly higher than DA levels in the striata of mice receiving MPTP only, thus indicating a degree of neuroprotection in this model of Parkinson's disease. The potential consequences on MAO activity of long term exposure to this compound need to be evaluated. Furthermore, there is evidence for the presence of other inhibitors in the tobacco leaf and smoke, including compounds with irreversible MAO inhibitory properties. Although there is no evidence to link the lowered activities of MAO to the lowered incidence of Parkinson's disease in smokers, the neuroprotective effects of TMN in the MPTP mouse model suggest that such a relationship is worthy of further evaluation.

2.
Ann N Y Acad Sci ; 939: 330-9, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11462789

ABSTRACT

Chronic (8- to 10-week) administration of the selective, potent, and irreversible monoamine oxidase B inhibitor (R)-deprenyl has been shown to increase the tyrosine hydroxylase immunoreactivity in the substantia nigra of mice that had been treated three days earlier with a neurotoxic dose of the parkinsonian-inducing agent 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). This reported rescuing of lesioned nigrostriatal cell bodies by (R)-deprenyl prompted us to investigate if this (R)-deprenyl treatment also could restore neostriatal dopamine levels that are depleted by MPTP. The results of these experiments show that long term (8 or 10 weeks) treatment with (R)-deprenyl beginning three days post MPTP administration did not result in restoration of depleted neostriatal dopamine levels in C57BL/6 mice. We conclude that, although (R)-deprenyl may rescue MPTP-injured nigrostriatal neurons, it does not lead to functional recovery of these neurons as measured by the restoration of neostriatal dopamine levels.


Subject(s)
Antiparkinson Agents/pharmacology , Dopamine/metabolism , Neostriatum/drug effects , Neurons/drug effects , Parkinsonian Disorders/metabolism , Selegiline/pharmacology , Animals , Antiparkinson Agents/therapeutic use , Cell Death/drug effects , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Neostriatum/injuries , Neostriatum/metabolism , Neurons/metabolism , Parkinsonian Disorders/drug therapy , Selegiline/therapeutic use
3.
J Neurosci ; 21(10): RC143, 2001 May 15.
Article in English | MEDLINE | ID: mdl-11319241

ABSTRACT

Recent epidemiological studies have established an association between the common consumption of coffee or other caffeinated beverages and a reduced risk of developing Parkinson's disease (PD). To explore the possibility that caffeine helps prevent the dopaminergic deficits characteristic of PD, we investigated the effects of caffeine and the adenosine receptor subtypes through which it may act in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) neurotoxin model of PD. Caffeine, at doses comparable to those of typical human exposure, attenuated MPTP-induced loss of striatal dopamine and dopamine transporter binding sites. The effects of caffeine were mimicked by several A(2A) antagonists (7-(2-phenylethyl)-5-amino-2-(2-furyl)-pyrazolo-[4,3-e]-1,2,4-triazolo[1,5-c]pyrimidine (SCH 58261), 3,7-dimethyl-1-propargylxanthine, and (E)-1,3-diethyl-8 (KW-6002)-(3,4-dimethoxystyryl)-7-methyl-3,7-dihydro-1H-purine-2,6-dione) (KW-6002) and by genetic inactivation of the A(2A) receptor, but not by A(1) receptor blockade with 8-cyclopentyl-1,3-dipropylxanthine, suggesting that caffeine attenuates MPTP toxicity by A(2A) receptor blockade. These data establish a potential neural basis for the inverse association of caffeine with the development of PD, and they enhance the potential of A(2A) antagonists as a novel treatment for this neurodegenerative disease.


Subject(s)
Caffeine/administration & dosage , Neuroprotective Agents/administration & dosage , Parkinsonian Disorders/drug therapy , Purinergic P1 Receptor Antagonists , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/antagonists & inhibitors , 3,4-Dihydroxyphenylacetic Acid/metabolism , Animals , Catechols/metabolism , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Corpus Striatum/pathology , Disease Models, Animal , Dopamine/metabolism , Dose-Response Relationship, Drug , Immunity, Innate/genetics , Injections, Intraperitoneal , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/genetics , Purines/administration & dosage , Pyrimidines/administration & dosage , Receptor, Adenosine A2A , Receptors, Purinergic P1/deficiency , Receptors, Purinergic P1/genetics , Theobromine/administration & dosage , Theobromine/analogs & derivatives , Triazoles/administration & dosage , Xanthines/administration & dosage
4.
Chem Res Toxicol ; 14(5): 523-7, 2001 May.
Article in English | MEDLINE | ID: mdl-11368550

ABSTRACT

Epidemiological evidence suggests a lower incidence of Parkinson's disease in smokers than in nonsmokers. This evidence, together with the lower levels of brain monoamine oxidase (MAO) activity in smokers and the potential neuroprotective properties of MAO inhibitors, prompted studies which led to the isolation and characterization of 2,3,6-trimethyl-1,4-naphthoquinone (TMN), an MAO-A and MAO-B inhibitor which is present in tobacco and tobacco smoke. Results of experiments reported here provide evidence that this compound protects against the MPTP-mediated depletion of neostriatal dopamine levels in the C57BL/6 mouse. These results support the hypothesis that the inhibition of MAO by constituents of tobacco smoke may be related to the decreased incidence of Parkinson's disease in smokers.


Subject(s)
Monoamine Oxidase/drug effects , Naphthoquinones/isolation & purification , Naphthoquinones/pharmacology , Neuroprotective Agents/isolation & purification , Neuroprotective Agents/pharmacology , Parkinsonian Disorders/prevention & control , Animals , Brain/metabolism , Disease Models, Animal , Dopamine/metabolism , Mice , Mice, Inbred C57BL , Monoamine Oxidase/metabolism , Monoamine Oxidase Inhibitors/isolation & purification , Monoamine Oxidase Inhibitors/metabolism , Monoamine Oxidase Inhibitors/pharmacology , Naphthoquinones/metabolism , Neostriatum/metabolism , Neuroprotective Agents/metabolism , Parkinsonian Disorders/chemically induced , Plant Extracts , Plants, Toxic , Nicotiana
5.
Exp Neurol ; 168(2): 434-6, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11259132

ABSTRACT

C. Thiffault, L. Lamarre-Théroux, R. Quirion, and J. Poirier (1997, Mol. Brain Res. 44: 238-244) recently reported that chronic treatment of young (12 week old) C57BL/6 mice with (R)-deprenyl, a mechanism-based inactivator of monoamine oxidase B (MAO-B), leads to a more than fourfold increase in neostriatal dopamine levels. Such an effect could complicate the interpretation of results obtained from mechanistic studies designed to evaluate the putative neuroprotective effects of (R)-deprenyl in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-lesioned mice. In contrast to the results of Thiffault et al., we have found that neostriatal dopamine levels in mature (32 week old) C57BL/6 mice were unaltered by chronic (R)-deprenyl treatment even though brain monoamine oxidase B activity was reduced by more than 80%. Neostriatal dopamine levels also were unaltered in both young and mature mice when the (R)-deprenyl treatment period was doubled compared to that reported by Thiffault et al. Consequently, studies on the putative neuroprotective properties of (R)-deprenyl in MPTP-lesioned mice are unlikely to be complicated by the possibility that inhibition of MAO-B alone will lead to an increase in neostriatal dopamine levels.


Subject(s)
Corpus Striatum/drug effects , Dopamine/metabolism , Monoamine Oxidase Inhibitors/administration & dosage , Monoamine Oxidase/drug effects , Selegiline/administration & dosage , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Animals , Corpus Striatum/injuries , Corpus Striatum/metabolism , Dopamine Agents , Male , Mice , Mice, Inbred C57BL , Neuroprotective Agents/administration & dosage , Parkinsonian Disorders
6.
Chem Res Toxicol ; 13(5): 336-41, 2000 May.
Article in English | MEDLINE | ID: mdl-10813649

ABSTRACT

This paper reports the results of studies on the in vivo metabolic fate of the tobacco alkaloid 1-methyl-2-(3-pyridinyl)pyrrole (beta-nicotyrine) in New Zealand white rabbits. Two previously characterized metabolites, 5-hydroxy-1-methyl-5-(3-pyridinyl)-2-pyrrolidinone (5-hydroxycotinine) and 2-hydroxy-1-methyl-5-(3-pyridinyl)-3-pyrrolin-2-one, were present in low concentrations in the urine of the treated animals. The major urinary metabolite of beta-nicotyrine was identified as cis-3'-hydroxy-1-methyl-5-(3-pyridinyl)-2-pyrrolidinone (cis-3'-hydroxycotinine), the diastereoisomer of the major urinary metabolite of (S)-nicotine. The pathway leading to cis-3'-hydroxycotinine is proposed to proceed via autoxidation of 2-hydroxy-1-methyl-5-(3-pyridinyl)pyrrole, a postulated cytochrome P450-generated metabolite of beta-nicotyrine, followed by reduction of the carbon-carbon double bond present in the resulting 3-hydroxy-3-pyrrolin-2-one species. This proposal is supported by the in vivo biotransformation of 2-acetoxy-1-methyl-5-(3-pyridinyl)pyrrole, a latent form of the putative hydroxypyrrole intermediate, to cis-3'-hydroxycotinine. The in vivo conversion of 5-hydroxy-1-methyl-5-(3-pyridinyl)-3-pyrrolin-2-one to 5-hydroxycotinine is offered as evidence that supports the proposed reduction step.


Subject(s)
Insecticides/pharmacokinetics , Plants , Pyridines/pharmacokinetics , Animals , Biotransformation , Chromatography, High Pressure Liquid , Gas Chromatography-Mass Spectrometry , Inactivation, Metabolic , Male , Mice , Mice, Inbred ICR , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Rabbits
7.
Neurotox Res ; 1(4): 261-9, 2000 Apr.
Article in English | MEDLINE | ID: mdl-12835094

ABSTRACT

Several laboratories recently have reported that melatonin may possess neuroprotective properties. The present paper presents the results of our studies on the long term in vivo neuroprotective effects of melatonin in a well-defined neurotoxicity model using 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) in the C57BL/6 mouse. MPTP is bioactivated by brain monoamine oxidase B (MAO-B) to its neurotoxic pyridinium metabolite 1-methyl-4-phenylpyridinium (MPP(+)) which destroys dopaminergic nerve terminals leading to the depletion of neostriatal dopamine (DA) and 3,4-dihydroxyphenylacetic acid (DOPAC). Our initial study compared striatal DA and DOPAC levels in MPTP-only-treated animals and animals treated with melatonin 30 min prior to and 3 times hourly post-MPTP. DA/DOPAC levels measured 7 days after MPTP were similar in both groups. A second study was designed to address the possibility that melatonin cleared from the brain prior to MPP(+). Animals, that had been administered the same regimen of melatonin as in the first study plus a fourth post-MPTP melatonin dose, were maintained on melatonin in drinking water until 5 days post-MPTP. Striatal DA/DOPAC levels of these melatonin-plus-MPTP treated animals also were the same as the MPTP-only-treated animals. In vitro studies confirmed that melatonin is not an inhibitor of MAO-B. These data demonstrate that melatonin does not have any significant protective effects against the long-term striatal DA and DOPAC depletion induced by MPTP in the C57BL/6 mouse.

8.
Neurobiology (Bp) ; 7(2): 135-49, 1999.
Article in English | MEDLINE | ID: mdl-10591048

ABSTRACT

The neurodegenerative properties of the parkinsonian inducing agent 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) are thought to result from inhibition of complex I of the mitochondrial respiratory chain by the monoamine oxidase-B (MAO-B) generated 1-methyl-4-phenylpyridinium metabolite MPP+. 7-Nitroindazole (7-NI) both a reversible MAO-B inhibitor and a neuronal nitric oxide synthase (nNOS) inhibitor, and (R)-deprenyl a potent MAO-B inactivator, provide neuroprotection in the C57BL/6 mouse model of MPTP neurotoxicity. The results reported here demonstrate the complexities of the effects of 7-NI in this model and examine the possibility of other mechanisms of neuroprotection by (R)-deprenyl.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Corpus Striatum/drug effects , Indazoles/pharmacology , Neuroprotective Agents/pharmacology , Neurotoxins/pharmacology , Selegiline/pharmacology , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/metabolism , 1-Methyl-4-phenylpyridinium/metabolism , 3,4-Dihydroxyphenylacetic Acid/metabolism , Animals , Cattle , Corpus Striatum/metabolism , Dopamine/metabolism , Male , Mice , Mice, Inbred C57BL , Monoamine Oxidase/drug effects , Monoamine Oxidase/metabolism , Neurotoxins/metabolism
9.
J Pharmacol Exp Ther ; 291(2): 856-64, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10525109

ABSTRACT

In an attempt to provide a better understanding of the scope and limitations of animal models used in some drug development programs and to further our understanding of potential metabolic bioactivation reactions, we have undertaken studies to profile the monoamine oxidase A and B (MAO-A and -B, respectively) activities in liver and brain mitochondrial preparations obtained from a variety of species using a series of 1-methyl-4-aryl-1,2,3, 6-tetrahydropyridinyl substrates. Mitochondrial preparations were incubated with substrates at 37 degrees C in the presence or absence of clorgyline, (R)-deprenyl, or a mixture of these two propargylamines to inhibit MAO-A, MAO-B, or both enzymes. The rates of formation of the corresponding dihydropyridinium metabolites were estimated spectrophotometrically. MAO-B was found to be the principal enzyme present in all tissues. Human liver displayed more MAO-A activity than the liver of any other species studied; subhuman primates displayed little or no detectable MAO-A activity. The properties of the preparations from rat liver were most similar to those from human liver with respect to the MAO-A/MAO-B ratios and the kinetic parameters of the four substrates used to profile enzymatic activity. The kinetic properties of mitochondrial preparations from bovine liver, a commonly used source of purified MAO-B preparations, were consistently different from all of the other species studied. The mitochondrial preparations from rabbit brain and liver also were unusual in that they displayed relatively low MAO activities. Additionally, these enzyme activities were considerably less susceptible to inhibition by clorgyline and (R)-deprenyl. Finally, an exceptionally low MAO-B liver/brain V(max)/K(m) ratio was observed with the mitochondria obtained from the C57BL/6 mouse, an effect that may contribute to the susceptibility of this strain to the toxic effects of the parkinsonian-inducing neurotoxin 1-methyl-4-phenyl-1,2,3, 6-tetrahydropyridine.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/metabolism , Mitochondria/metabolism , Monoamine Oxidase/physiology , Animals , Brain/metabolism , Cattle , Clorgyline/pharmacology , Dogs , Humans , Macaca fascicularis , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mitochondria, Liver/metabolism , Monoamine Oxidase/classification , Monoamine Oxidase Inhibitors/pharmacology , Papio , Rabbits , Rats , Rats, Sprague-Dawley , Selegiline/pharmacology , Species Specificity , Spectrophotometry
10.
Pol J Pharmacol ; 51(1): 31-8, 1999.
Article in English | MEDLINE | ID: mdl-10389142

ABSTRACT

The pyridinium metabolites formed in the MAO-B catalyzed oxidation of 1-methyl-4-substituted-1,2,3,6-tetrahydropyridinyl derivatives, such as the parkinsonian inducing agent 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), cause the selective degeneration of nigrostriatal neurons, presumably by inhibition of mitochondrial respiration and depletion of ATP stores. The possibility that other partially oxidized piperidinyl derivatives also may be biotransformed to toxic pyridinium metabolites has led us to examine the metabolic fate of the neuroleptic agent haloperidol (HP) and its tetrahydropyridinyl dehydration product 4-(4-chlorophenyl)-1[4-(4-fluorophenyl)-4-oxobutyl]- 1,2,3,6-tetrahydropyridine (HPTP). In vitro metabolic studies employing tissue preparations isolated from rodents, baboons and humans have documented that cytochrome P4503A enzymes catalyze the biotransformation of both HP and HPTP to yield the corresponding pyridinium metabolite HPP+. An analogous biotransformation profile has been observed with "reduced haloperidol" (RHP), an abundant, circulating metabolite of HP formed by the stereospecific reduction of the benzoyl carbonyl group of HP. In vivo studies also have documented these pathways in humans, baboons and rodents. Although both HPP+ and RHPP+ are found in the urine and plasma of HP treated patients and HP or HPTP treated baboons, attempts to identify an MPTP-type lesion in baboons following long-term treatment with HPTP have failed. On the other hand, evidence for a lesion of the nucleus basalis of Meynert has been obtained. Additionally, the urinary excretion of abnormal organic acids and acylcarnitine conjugates suggests that HP and/or metabolites derived from HP interfere with energy production pathways.


Subject(s)
Amines/metabolism , Haloperidol/metabolism , Nervous System Diseases/chemically induced , Neurotoxins/biosynthesis , Pyridinium Compounds/metabolism , Animals , Biotransformation , Cytochrome P-450 Enzyme System/metabolism , Dopamine Antagonists/pharmacology , Haloperidol/pharmacology , Humans , MPTP Poisoning , Structure-Activity Relationship
11.
Chem Res Toxicol ; 12(6): 508-12, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10368313

ABSTRACT

Previous studies have established that the tobacco alkaloid 1-methyl-2-(3-pyridyl)pyrrole (beta-nicotyrine) is biotransformed by rabbit lung and liver microsomal preparations to an equilibrium mixture of the corresponding 3- and 4-pyrrolin-2-ones. Autoxidation of these pyrrolin-2-ones generates the chemically stable 5-hydroxy-5-(3-pyridinyl)-3-pyrrolin-2-one. This paper summarizes efforts to document more completely the pathway leading to this hydroxypyrrolinone. Chemical and spectroscopic evidence implicates the 2-hydroxy-1-methyl-5-(3-pyridinyl)pyrrole (2-hydroxy-beta-nicotyrine) as the key intermediate in this reaction pathway. Of potential toxicological interest is the detection of radical species derived from the autoxidation of this compound.


Subject(s)
Insecticides/metabolism , Nicotiana/metabolism , Plants, Toxic , Pyridines/metabolism , Pyrroles/metabolism , Animals , Free Radicals , Oxidation-Reduction , Rabbits
12.
Bioorg Med Chem ; 6(12): 2531-9, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9925309

ABSTRACT

NADPH supplemented rat liver microsomal enzyme preparations catalyze the conversion of 1-cyclopropyl4-phenyl-1,2,3,6-tetrahydropyridine to the p-hydroxyphenyl (low yield), descyclopropyl (high yield) and 2,3-dihydropyridinium and, subsequently, pyridinium (intermediary yield) metabolites. When the methine proton of the cyclopropyl group was replaced with a deuteron, a normal deuterium isotope effect (1.4) was observed on the formation of the decyclopropylated metabolite and an inverse isotope effect (0.6) on the dihydropyridinium metabolite. A larger deuterium isotope effect (3.6) was observed on the ring alpha-carbon oxidation pathway with the 2,2,6,6-d4 analogue as substrate. These results and the observation that the ratios of the rates of these two alpha-carbon oxidation pathways are independent of initial substrate concentrations suggest that both pathways are catalyzed by the same active site of one form of P450. These transformations are discussed in terms of metabolic pathways that have been proposed for the cytochrome P450 catalyzed alpha-carbon oxidation of amines.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/analogs & derivatives , Cytochrome P-450 Enzyme System/metabolism , Microsomes, Liver/enzymology , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/chemistry , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacokinetics , Animals , Biotransformation , Deuterium , Kinetics , Male , Models, Chemical , Models, Molecular , Molecular Conformation , Molecular Structure , NADP/metabolism , Oxidation-Reduction , Radioisotope Dilution Technique , Rats , Rats, Sprague-Dawley , Substrate Specificity
13.
J Neurochem ; 69(4): 1771-3, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9326308

ABSTRACT

The ability of 7-nitroindazole (7-NI) to protect against MPTP-induced neurotoxicity has been attributed to its inhibition of neuronal nitric oxide synthase. In the present study, 7-NI was found to counteract almost completely striatal dopamine depletion caused by a single subcutaneus injection of 20 mg/kg MPTP in mice. This effect, however, was accompanied by a significant reduction in the striatal levels of MPP+, the toxic metabolite generated via monoamine oxidase B-catalyzed MPTP oxidation. In the presence of 7-NI, a dose of 40 mg/kg MPTP produced MPP concentrations similar to those measured after treatment with 20 mg/kg MPTP alone. A comparison of neurotoxicity in these two experimental conditions (i.e., mice treated with 20 mg/kg alone versus 40 mg/kg MPTP plus 7-NI) revealed only a slight (20%), but statistically significant, protection of dopamine depletion with 7-NI. These data indicate that the mechanism by which 7-NI counteracts MPTP neurotoxicity in mice is not due solely to inhibition of neuronal nitric oxide synthase, but involves a reduction in MPP+ formation.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Enzyme Inhibitors/pharmacology , Indazoles/pharmacology , Monoamine Oxidase Inhibitors/pharmacology , Neuroprotective Agents/pharmacology , Neurotoxins/pharmacology , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/antagonists & inhibitors , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/metabolism , 1-Methyl-4-phenylpyridinium/metabolism , Animals , Corpus Striatum/metabolism , Male , Mice , Mice, Inbred C57BL , Neurons/enzymology , Neurotoxins/antagonists & inhibitors , Nitric Oxide Synthase/antagonists & inhibitors
14.
Chem Res Toxicol ; 10(9): 924-40, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9305573

ABSTRACT

A major theme explored in this review is the MAO-and cytochrome P450-catalyzed alpha-carbon oxidations of selected cyclic tertiary amines to give iminium metabolites that undergo further chemical modifications to form known or potentially toxic products. The most dramatic illustration of this type of bioactivation process is the conversion of the parkinsonian-inducing neurotoxin MPTP (23) by brain MAO-B to the iminium (dihydropyridinium) metabolite 24 which is oxidized further to the pyridinium species MPP+ (25). The selective destruction of nigrostriatal neurons by MPP+ is dependent on a unique sequence of events (transport into the nerve terminals by the dopamine transporter, localization in the inner mitochondrial membrane by electromotive forces, and inhibition of complex I of the mitochondrial electron transport chain) that, fortunately, are unlikely to be encountered with many substances. A second example of a well-documented metabolic bioactivation sequence involves the highly toxic pyrrolizidine alkaloids (102). These compounds undergo cytochrome P450-catalyzed alpha-carbon oxidation which converts the 3-pyrrolinyl moiety present in the parent alkaloids into a pyrrolyl-containing metabolite (105). The presence of labile functional groups results in the spontaneous conversion of 105 to reactive electrophilic products (106 and 108) that undergo Michael addition reactions with nucleophiles on biomacromolecules leading to a variety of toxic outcomes. Less clearly defined are the potential contributions to neurodegenerative processes that may be mediated by low-level, long term exposure to less potent toxins. Examples of potential proneurotoxins are the endogenously formed tetrahydroisoquinolines (such as 40-50) and tetrahydro-beta-carbolines (such as 54) that may be biotransformed to neurotoxic isoquinolinium (such as 51) and beta-carbolinium (such as 52) species in the brain. A similar argument can be made for 4-piperidinols (compounds that are at the same oxidation state as the tetrahydropyridines) which may be metabolized via iminium intermediates to amino enols that spontaneously convert to dihydropyridinium species and hence to pyridinium metabolites (67-->68-->69-->70-->71, Scheme 10). This type of reaction sequence has been well documented with the parkinsonian-inducing neuroleptic agent haloperidol (72) which is metabolized in humans, baboons, and rodents to the pyridinium species HPP+ (75), a potent inhibitor of mitochondrial respiration. Finally, an appreciation of the alpha-carbon oxidations of fully reduced azacycles such as (S)-nicotine (61) and phencyclidine (82) to chemically reactive metabolites that form covalent adducts with proteins, including the enzymes that are responsible for their formation, may prove of toxicological importance when attempting to account for the effects of chronic abuse of these potent drugs.1


Subject(s)
Amines/metabolism , Aza Compounds/metabolism , Animals , Biotransformation , Humans , Oxidation-Reduction
15.
Chem Res Toxicol ; 10(4): 364-8, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9114971

ABSTRACT

The neurodegenerative properties of the parkinsonian inducing agent 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) are thought to result from inhibition of complex I of the mitochondrial respiratory chain by the monoamine oxidase-B (MAO-B) generated 1-methyl-4-phenylpyridinium metabolite MPP+. Treatment with 7-nitroindazole (7-NI) protects rodents and baboons against MPTP's neurotoxicity, presumably as a consequence of its inhibition of neuronal nitric oxide synthase (nNOS). The results reported in the present communication, while not in conflict with the proposed role of nNOS, raise the possibility that the inhibition of MAO-B by 7-NI also may contribute to the observed neuroprotection.


Subject(s)
Enzyme Inhibitors/pharmacology , Indazoles/pharmacology , Monoamine Oxidase/metabolism , Neurons/drug effects , Nitric Oxide Synthase/antagonists & inhibitors , Animals , Catalysis , Cattle , Kinetics , Male , Mice , Mice, Inbred C57BL , Neurons/enzymology , Oxidation-Reduction
17.
J Med Chem ; 39(24): 4756-61, 1996 Nov 22.
Article in English | MEDLINE | ID: mdl-8941389

ABSTRACT

The results of previous studies have established that the monoamine oxidase-catalyzed oxidation of 1-methyl-1,2,3,6-tetrahydropyridyl derivatives bearing heteroatom substituents at C-4 generates 2,3-dihydropyridinium intermediates that undergo spontaneous hydrolysis to release the C-4 substituent and form the amino enone 1-methyl-2,3-dihydro-4-pyridone. We have attempted to adapt this metabolic pathway to the preparation of amine-containing prodrugs that may target the central nervous system which is rich in monoamine oxidase A and B. In this paper we report the synthesis and the in vitro and in vivo metabolic fate of the tetrahydropyridyl carbamate derivatives which are designed to release (S)- and (R)-nordeprenyl. These carbamates are selective monoamine oxidase A substrates. An ex vivo assay has shown that the R-enantiomer is an effective and selective inhibitor of brain mitochondrial monoamine oxidase B.


Subject(s)
Carbamates/chemical synthesis , Monoamine Oxidase Inhibitors/chemical synthesis , Pyridines/chemical synthesis , Animals , Brain/drug effects , Brain/enzymology , Carbamates/pharmacology , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Humans , Kinetics , Magnetic Resonance Spectroscopy , Male , Mice , Mice, Inbred Strains , Mitochondria/enzymology , Molecular Conformation , Monoamine Oxidase/metabolism , Monoamine Oxidase Inhibitors/pharmacology , Placenta/enzymology , Prodrugs/metabolism , Pyridines/pharmacology , Substrate Specificity
18.
Chem Res Toxicol ; 9(6): 1013-22, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8870990

ABSTRACT

The monoamine oxidase B (MAO-B) catalyzed oxidation of amines has been proposed to proceed via a polar pathway, an initial single-electron transfer pathway and an initial hydrogen atom transfer pathway. Results from previous studies on selected N-cyclopropyl-4-substituted-1,2,3,6-tetrahydropyridine derivatives have led us to consider a mechanism for these cyclic tertiary allylamines which may not necessarily involve the aminyl radical cation as required by an initial single-electron transfer step. The studies summarized in this paper were undertaken to explore further the structural features that determine the MAO-B substrate and/or inactivator properties of various 1,4-disubstituted tetrahydropyridine derivatives. We report here the results of our studies on the synthesis and MAO-B catalyzed oxidation of 1-methyl- and 1-cyclopropyl-1,2,3,6-tetrahydropyridine derivatives bearing a variety of heteroaromatic groups at C-4. All of the N-cyclopropyltetrahydropyridine analogs were time and concentration dependent inhibitors of MAO-B while all of the N-methyltetrahydropyridine analogs and the N-cyclopropyl-4-(1-methyl-2-pyrryl)tetrahydropyridine analog were substrates. The substrate properties (Kcat/KM) covered a range of 6 to 1800 min-1 mM-1 while the range for the inactivator properties for which Kinact/KI values could be obtained was 0.1-1.0 min-1 mM-1. The partition ratios for the N-cyclopropyl analogs varied from 4 to 17 except for the 4-(1-methyl-2-pyrryl) analog, which had a partition ratio of 400. These results are discussed in terms of the putative allylic radical intermediate and in the context of the hydrogen atom transfer and single-electron transfer based mechanisms.


Subject(s)
Monoamine Oxidase/chemistry , Pyridines/chemistry , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/analogs & derivatives , Animals , Catalysis , Cattle , Kinetics , Monoamine Oxidase Inhibitors/chemistry , Oxidation-Reduction , Pyridines/chemical synthesis , Substrate Specificity
19.
J Pharmacol Exp Ther ; 270(2): 822-30, 1994 Aug.
Article in English | MEDLINE | ID: mdl-8071874

ABSTRACT

It is now generally accepted that the nigrostriatal degenerative properties of the parkinsonian-inducing agent 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine are mediated by the brain monoamine oxidase B generated 1-methyl-4-phenylpyridinium metabolite (MPP+). In this article, the results are described of ongoing efforts to evaluate the MPP(+)-type neurotoxic potential of the haloperidol (HP)-derived pyridinium metabolite HPP+, a 1,4-disubstituted structural analog of MPP+, which is formed in humans and rats treated with HP. Previous studies in the rat have shown that intrastriatal perfusion of HPP+ leads to the irreversible depletion of striatal dopamine and serotonin. Furthermore, HPP+ was a potent inhibitor of NADH-supported mitochondrial respiration. This article reports that HPP+ also is toxic to dopaminergic and serotonergic neurons in cultures of embryonic mesencephalic cells, as measured by loss of the ability of exposed cells to accumulate tritium-labeled dopamine and serotonin and by immunochemical staining techniques. HPP+ also inhibited the uptake of these labeled neurotransmitters by synaptosomes prepared from mouse neostriata (dopamine) and cortical tissues (serotonin). Because HP is unlikely to be a substrate for brain monoamine oxidase B, the production and accumulation of HPP+ in the brain is probably not comparable to that of MPP+. On the other hand, chronic exposure to HP could result in brain levels of this lipophilic quaternary pyridinium species that might coincide with the late-appearing tardive dyskinesias that are observed in some HP-treated patients months and, more often, years after the initiation of HP therapy.


Subject(s)
1-Methyl-4-phenylpyridinium/toxicity , Haloperidol/analogs & derivatives , Animals , Cells, Cultured , Dopamine/metabolism , Female , Haloperidol/metabolism , Pregnancy , Rats , Rats, Sprague-Dawley , Serotonin/metabolism , Synaptosomes/drug effects , Synaptosomes/metabolism
20.
Chem Res Toxicol ; 7(3): 281-5, 1994.
Article in English | MEDLINE | ID: mdl-8075357

ABSTRACT

The neuroleptic agent haloperidol (HP) is biotransformed in humans to a pyridinium metabolite, HPP+, that displays neurotoxic properties resembling those of the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-derived neurotoxic pyridinium metabolite MPP+. We report here that HP and its tetrahydropyridine dehydration product 4-(4-chlorophenyl)-1-[4-(4-fluorophenyl)-4-oxobutyl]-1,2,3,6- tetrahydropyridine (HPTP) are metabolized in vivo by the MPTP-susceptible C57BL/6 mouse to several pyridinium metabolites including HPP+ and the 4-(4-chlorophenyl)-1-[4-(4-fluorophenyl)-4-hydroxybutyl]pyridinium species RHPP+, the pyridinium species corresponding to reduced haloperidol (RHP), a major circulating metabolite of HP. Atmospheric pressure ion-spray (API) mass spectral data also suggest the formation of fluorophenyl ring-hydroxylated derivatives of these two pyridinium metabolites. Furthermore, HPLC tracings reveal the presence of HPP+, RHPP+, and two phenolic pyridinium metabolites in brain tissue extracts of HPTP, but not HP, treated mice. The neurotoxic potential of MPTP-type pyridinium species suggests that these metabolites may contribute to some of the neurological disorders observed in humans undergoing chronic HP treatment.


Subject(s)
Haloperidol/pharmacokinetics , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/metabolism , Animals , Biotransformation , Brain/metabolism , Chromatography, High Pressure Liquid , Haloperidol/analogs & derivatives , Haloperidol/urine , Male , Mice , Mice, Inbred C57BL , Neurotoxins/metabolism , Spectrometry, Fluorescence
SELECTION OF CITATIONS
SEARCH DETAIL
...