Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
Add more filters










Publication year range
1.
Neurotox Res ; 7(4): 251-63, 2005.
Article in English | MEDLINE | ID: mdl-16179262

ABSTRACT

A number of studies have implicated the interactions of the excitatory amino acid L-glutamate (Glu) with its ionotropic and metabotropic receptors as important components of the mechanism underlying the dopaminergic neurotoxicity of 1-methyl-4-phenylpyridinium [MPP(+)]. Furthermore, microdialysis experiments have demonstrated that perfusion of relatively high concentrations of MPP(+) into the rat striatum evoke a delayed, massive release of Glu. Interestingly, perfusion of MPP(+) also mediates a similar release of glutathione (GSH). Together, these observations raise the possibility that the rise of extracellular Glu mediated by MPP(+) may be the result of hydrolysis of released GSH by gamma-glutamyl transpeptidase (gamma-GT). In the present investigation it is demonstrated that perfusions of solutions of 0.7 and 1.3 mM MPP(+) dissolved in artificial cerebrospinal fluid into the rat striatum evoke neurotoxic damage to dopaminergic terminals, assessed by both a two-day test/challenge procedure and tyrosine hydroxylase immunoreactivity, but without the release of Glu. Perfusions of 2.5 mM MPP(+) cause more extensive dopaminergic neurotoxicity and a dose-dependent release of Glu. However, neither this release of Glu nor MPP(+)-induced dopaminergic neurotoxicity are blocked by the irreversible gamma-GT inhibitor acivicin. Together, these observations indicate that a rise of extracellular levels of Glu is not essential for the dopaminergic neurotoxicity of MPP(+). Furthermore, the rise of extracellular Glu caused by perfusion of 2.5 mM MPP(+) is not the result of the gamma-GT-mediated hydrolysis of released GSH. It is possible that the rise of extracellular levels of Glu, L-aspartate, L-glycine and L-taurine evoked by perfusions of 2.5 mM MPP(+) into the rat striatum may reflect, at least in part, the release of these amino acids from astrocytes.


Subject(s)
1-Methyl-4-phenylpyridinium/toxicity , Dopamine Agents/toxicity , Extracellular Space/metabolism , Glutamic Acid/metabolism , Glutathione/metabolism , Neostriatum/metabolism , Neurotoxicity Syndromes/metabolism , Amino Acids/metabolism , Animals , Chromatography, High Pressure Liquid , Dopamine/metabolism , Electrochemistry , Enzyme Inhibitors/pharmacology , Extracellular Space/drug effects , Immunohistochemistry , Isoxazoles/pharmacology , Male , Microdialysis , Neostriatum/drug effects , Rats , Rats, Sprague-Dawley , Serotonin/metabolism , gamma-Glutamyltransferase/antagonists & inhibitors , gamma-Glutamyltransferase/metabolism
2.
Chem Res Toxicol ; 14(9): 1184-92, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11559032

ABSTRACT

Tryptamine-4,5-dione (T-4,5-D) is formed as a result of oxidation of 5-hydroxytryptamine by superoxide (O(2)(-)(*), nitric oxide (NO*), and peroxynitrite (ONOO(-)). T-4,5-D rapidly inactivates tryptophan hydroxylase (TPH), derived from rat brain, probably as a result of covalent modification of active site cysteine residues. The activity of TPH exposed to T-4,5-D cannot be restored by anaerobic reduction with dithiothreitol (DTT) and ferrous iron (Fe(2+)) indicating that the inactivation is irreversible. 7-S-Glutathionyl-tryptamine-4,5-dione, formed by the rapid reaction between T-4,5-D and glutathione, also inhibits TPH but in this case the activity is restored by anaerobic reduction with DTT/Fe(2+). The results of this investigation may be relevant to the initial reversible and subsequent irreversible inactivation of TPH evoked by methamphetamine and 3,4-methylenedioxymethamphetamine.


Subject(s)
Central Nervous System Stimulants/toxicity , Indolequinones , Methamphetamine/toxicity , Tryptamines/pharmacology , Tryptophan Hydroxylase/drug effects , Tryptophan Hydroxylase/metabolism , Animals , Brain/enzymology , Brain/physiology , Central Nervous System Stimulants/pharmacology , Chromatography, High Pressure Liquid , Male , Methamphetamine/pharmacology , Oxidants , Oxidation-Reduction , Rats , Rats, Sprague-Dawley , Tryptamines/chemistry
3.
J Pharm Biomed Anal ; 25(2): 181-9, 2001 May.
Article in English | MEDLINE | ID: mdl-11275426

ABSTRACT

The oxidation chemistry of 3',4'-deoxynorlaudanosoline carboxylic acid, a tetrahydroisoquinoline alkaloid, has been studied by electrochemical approaches. Four reaction products were isolated by semi-preparative high performance liquid chromatography and identified structurally by nuclear magnetic resonance, mass spectrometry, ultraviolet-visible spectrophotometry and electrochemistry studies. An oxidation mechanism was proposed.


Subject(s)
Alkaloids/isolation & purification , Tetrahydropapaveroline/chemistry , Chromatography, High Pressure Liquid , Electrochemistry , Hydrogen-Ion Concentration , Magnetic Resonance Spectroscopy , Oxidation-Reduction , Spectrometry, Mass, Fast Atom Bombardment , Tetrahydropapaveroline/analogs & derivatives , Tetrahydropapaveroline/isolation & purification
4.
J Neural Transm (Vienna) ; 108(12): 1363-74, 2001.
Article in English | MEDLINE | ID: mdl-11810401

ABSTRACT

The principal neuropathological feature of Parkinson's disease is the degeneration of melanized dopamine neurons in the substantia nigra pars compacta (SNc). Characteristic pathobiochemical changes in the parkinsonian SNc include a fall of both dopamine (DA) and glutathione levels (GSH), increased activity of gamma-glutamyl transpeptidase, a key enzyme involved in the degradation of GSH to L-cysteine (CySH), together with evidence for elevated intraneuronal superoxide (O2-*), nitric oxide (NO.) and thence peroxynitrite (ONOO-) generation, and accelerated DA oxidation as indicated by a large rise of the 5-S-cysteinyldopamine (5-S-CyS-DA)/DA concentration ratio. The latter effect is consistent with an increased rate of DA oxidation by O2-* and ONOO- forming DA-o-quinone which reacts with CySH forming 5-S-CyS-DA. However, 5-S-CyS-DA is readily further oxidized to 7-(2-aminoethyl)-3,4-dihydro-5-hydroxy-2H-1,4-benzothiazine-3-carboxylic acid (DHBT-1). Previous studies have demonstrated that DHBT-1 is rapidly accumulated by isolated intact rat brain mitochondria and selectively inhibits complex I respiration and the alpha-ketoglutarate dehydrogenase (alpha-KGDH) complex. In this study it is demonstrated that DHBT-1 also inhibits the pyruvate dehydrogenase complex (PDHC). The mechanism underlying the inhibition of all of these enzyme complexes involves bioactivation of intramitochondrial DHBT-1 by oxidation to highly electrophilic metabolites that covalently bind to active site cysteine residues. Thus, oxidative metabolites of intraneuronal 5-S-CyS-DA may contribute to impaired mitochondrial complex I and alpha-KGDH activities known to occur in the parkinsonian SNc and suggest that impaired PDHC evoked by the same metabolites may also occur in PD.


Subject(s)
Dopamine/analogs & derivatives , Dopamine/metabolism , Oxidative Stress/physiology , Parkinson Disease/enzymology , Pyruvate Dehydrogenase Complex/metabolism , Substantia Nigra/enzymology , Thiazines/metabolism , Animals , Ascorbic Acid/pharmacology , Catalase/metabolism , Catalase/pharmacology , Energy Metabolism/drug effects , Energy Metabolism/physiology , Glutathione/metabolism , Glutathione/pharmacology , Intracellular Membranes/drug effects , Intracellular Membranes/metabolism , Male , Mitochondria/drug effects , Mitochondria/enzymology , Neurons/enzymology , Parkinson Disease/physiopathology , Rats , Rats, Sprague-Dawley , Substantia Nigra/physiopathology , Superoxide Dismutase/metabolism , Superoxide Dismutase/pharmacology , Thiazines/pharmacology
6.
J Neural Transm (Vienna) ; 107(8-9): 959-78, 2000.
Article in English | MEDLINE | ID: mdl-11041275

ABSTRACT

A characteristic change in the substantia nigra of Parkinson's disease patients is an apparent accelerated rate of dopamine oxidation as evidenced by an increased 5-S-cysteinyldopamine (5-S-CyS-DA) to dopamine ratio. However, 5-S-CyS-DA is more easily oxidized than dopamine to give 7-(2-aminoethyl)-3,4-dihydro-5-hydroxy-2H-1,4-benzothiazine-3-carboxylic acid (DHBT-1). Previous studies have demonstrated that DHBT-1 can be accumulated by intact rat brain mitochondria and inhibits complex I but not complex II respiration. In this study, it is shown that DHBT-1 also inhibits the alpha-ketoglutarate dehydrogenase complex (alpha-KGDH) but not cytochrome c oxidase (complex IV). The inhibition of alpha-KGDH is dependent on the oxidation of DHBT-1, catalyzed by an unknown constituent of the inner mitochondrial membrane, to an electrophilic o-quinone imine that covalently modifies active site sulfhydryl residues. The latter conclusion is based on the ability of > or = equimolar glutathione to block the inhibition of alpha-KGDH by DHBT-1, without altering its rate of mitochondrial membrane-catalyzed oxidation, by scavenging the electrophilic o-quinone intermediate forming glutathionyl conjugates which have been isolated and spectroscopically characterized. Activities of mitochondrial alpha-KGDH and complex I, but not other respiratory complexes, are decreased in the parkinsonian substantia nigra. Such changes together with evidence for accelerated dopamine oxidation, increased formation of 5-S-CyS-DA and the ease of oxidation of this conjugate to DHBT-1 which inhibits alpha-KGDH and complex I, without affecting other respiratory enzyme complexes, suggests that the latter putative metabolite might be an endotoxin that contributes to the alpha-KGDH and complex I defects in Parkinson's disease.


Subject(s)
Brain/enzymology , Dopamine/analogs & derivatives , Dopamine/metabolism , Ketoglutarate Dehydrogenase Complex/metabolism , Parkinson Disease/etiology , Parkinson Disease/metabolism , Animals , Antioxidants/metabolism , Catalase/metabolism , Dopamine/pharmacology , Electron Transport Complex IV/metabolism , Enzyme Inhibitors/pharmacology , Intracellular Membranes/enzymology , Ketoglutarate Dehydrogenase Complex/antagonists & inhibitors , Male , Mitochondria/enzymology , Oxidation-Reduction , Prostaglandin-Endoperoxide Synthases/metabolism , Rats , Rats, Sprague-Dawley , Superoxide Dismutase/metabolism , Thiazines/metabolism , Thiazines/pharmacology
7.
Chem Res Toxicol ; 13(8): 749-60, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10956063

ABSTRACT

The major initial product of the oxidation of norepinephrine (NE) in the presence of L-cysteine is 5-S-cysteinylnorepinephrine which is then further easily oxidized to the dihydrobenzothiazine (DHBT) 7-(1-hydroxy-2-aminoethyl)-3,4-dihydro-5-hydroxy-2H-1, 4-benzothiazine-3-carboxylic acid (DHBT-NE-1). When incubated with intact rat brain mitochondria, DHBT-NE-1 evokes rapid inhibition of complex I respiration without affecting complex II respiration. DHBT-NE-1 also evokes time- and concentration-dependent irreversible inhibition of NADH-coenzyme Q(1) (CoQ(1)) reductase, the pyruvate dehydrogenase complex (PDHC), and alpha-ketoglutarate dehydrogenase (alpha-KGDH) when incubated with frozen and thawed rat brain mitochondria (mitochondrial membranes). The time dependence of the inhibition of NADH-CoQ(1) reductase, PDHC, and alpha-KGDH by DHBT-NE-1 appears to be related to its oxidation, catalyzed by an unknown component of the inner mitochondrial membrane, to electrophilic intermediates which bind covalently to active site cysteinyl residues of these enzyme complexes. The latter conclusion is based on the ability of glutathione to block inhibition of NADH-CoQ(1) reductase, PDHC, and alpha-KGDH by scavenging electrophilic intermediates, generated by the mitochondrial membrane-catalyzed oxidation of DHBT-NE-1, forming glutathionyl conjugates, several of which have been isolated and spectroscopically identified. The possible implications of these results to the degeneration of neuromelanin-pigmented noradrenergic neurons in the locus ceruleus in Parkinson's disease are discussed.


Subject(s)
Enzyme Inhibitors/pharmacology , Mitochondria/drug effects , Multienzyme Complexes/drug effects , Norepinephrine/analogs & derivatives , Parkinson Disease/metabolism , Thiazines/pharmacology , Animals , Brain/drug effects , Brain/enzymology , Brain/metabolism , Chromatography, High Pressure Liquid , Electron Transport Complex III/antagonists & inhibitors , Electron Transport Complex III/drug effects , Enzyme Inhibitors/chemical synthesis , Ketoglutarate Dehydrogenase Complex/antagonists & inhibitors , Ketoglutarate Dehydrogenase Complex/drug effects , Male , Mitochondria/enzymology , Multienzyme Complexes/antagonists & inhibitors , Norepinephrine/metabolism , Oxidation-Reduction , Pyruvate Dehydrogenase Complex/antagonists & inhibitors , Pyruvate Dehydrogenase Complex/drug effects , Rats , Rats, Sprague-Dawley
8.
Chem Res Toxicol ; 13(3): 135-60, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10725110

ABSTRACT

Quinones represent a class of toxicological intermediates which can create a variety of hazardous effects in vivo, including acute cytotoxicity, immunotoxicity, and carcinogenesis. The mechanisms by which quinones cause these effects can be quite complex. Quinones are Michael acceptors, and cellular damage can occur through alkylation of crucial cellular proteins and/or DNA. Alternatively, quinones are highly redox active molecules which can redox cycle with their semiquinone radicals, leading to formation of reactive oxygen species (ROS), including superoxide, hydrogen peroxide, and ultimately the hydroxyl radical. Production of ROS can cause severe oxidative stress within cells through the formation of oxidized cellular macromolecules, including lipids, proteins, and DNA. Formation of oxidatively damaged bases such as 8-oxodeoxyguanosine has been associated with aging and carcinogenesis. Furthermore, ROS can activate a number of signaling pathways, including protein kinase C and RAS. This review explores the varied cytotoxic effects of quinones using specific examples, including quinones produced from benzene, polycyclic aromatic hydrocarbons, estrogens, and catecholamines. The evidence strongly suggests that the numerous mechanisms of quinone toxicity (i.e., alkylation vs oxidative stress) can be correlated with the known pathology of the parent compound(s).


Subject(s)
Quinones/toxicity , Alkylation , Animals , Benzene/metabolism , Catecholamines/metabolism , Estrogens/metabolism , Humans , Oxidative Stress , Polycyclic Aromatic Hydrocarbons/metabolism , Quinones/chemistry , Quinones/metabolism
9.
J Neurochem ; 73(4): 1683-95, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10501216

ABSTRACT

In this investigation, microdialysis has been used to study the effects of 1-methyl-4-phenylpyridinium (MPP+), an inhibitor of mitochondrial complex I and alpha-ketoglutarate dehydrogenase and the active metabolite of the dopaminergic neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), on extracellular concentrations of glutathione (GSH) and cysteine (CySH) in the rat striatum and substantia nigra (SN). During perfusion of a neurotoxic concentration of MPP+ (2.5 mM) into the rat striatum or SN, extracellular concentrations of GSH and CySH remain at basal levels (both approximately 2 microM). However, when the perfusion is discontinued, a massive but transient release of GSH occurs, peaking at 5,000% of basal levels in the striatum and 2,000% of basal levels in the SN. The release of GSH is followed by a slightly delayed and smaller elevation of extracellular concentrations of CySH that can be blocked by the gamma-glutamyl transpeptidase (gamma-GT) inhibitor acivicin. Low-molecular-weight iron and extracellular hydroxyl radical (OH*) have been implicated as participants in the mechanism underlying the dopaminergic neurotoxicity of MPTP/MPP+. During perfusion of Fe2+ (OH*) into the rat striatum and SN, extracellular levels of GSH also remain at basal levels. When perfusions of Fe2+ are discontinued, a massive transient release of GSH occurs followed by a delayed, small, but progressive elevation of extracellular CySH level that again can be blocked by acivicin. Previous investigators have noted that extracellular concentrations of the excitatory/excitotoxic amino acid glutamate increase dramatically when perfusions of neurotoxic concentrations of MPP+ are discontinued. This observation and the fact that MPTP/MPP+ causes the loss of nigrostriatal GSH without corresponding increases of glutathione disulfide (GSSG) and the results of the present investigation suggest that the release and gamma-GT/dipeptidase-mediated hydrolysis of GSH to glutamate, glycine, and CySH may be important factors involved with the degeneration of dopamine neurons. It is interesting that a very early event in the pathogenesis of Parkinson's disease is a massive loss of GSH in the SN pars compacta that is not accompanied by corresponding increases of GSSG levels. Based on the results of this and prior investigations, a new hypothesis is proposed that might contribute to an understanding of the mechanisms that underlie the degeneration of dopamine neurons evoked by MPTP/MPP+, other agents that impair neuronal energy metabolism, and Parkinson's disease.


Subject(s)
1-Methyl-4-phenylpyridinium/pharmacology , Corpus Striatum/metabolism , Glutathione/metabolism , Iron/pharmacology , Parkinson Disease/metabolism , Propionates/pharmacology , Substantia Nigra/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Animals , Corpus Striatum/drug effects , Cysteine/metabolism , Glutamic Acid/metabolism , Glycine/metabolism , Hydrolysis , Hydroxyl Radical/pharmacology , Kinetics , Male , Microdialysis , Mitochondria/drug effects , Mitochondria/metabolism , Models, Chemical , Nitro Compounds , Oxygen Consumption/drug effects , Perfusion , Rats , Rats, Sprague-Dawley , Substantia Nigra/drug effects
10.
Chem Res Toxicol ; 12(5): 429-36, 1999 May.
Article in English | MEDLINE | ID: mdl-10328753

ABSTRACT

The release and subsequent reuptake of 5-hydroxytryptamine (5-HT) and cytoplasmic superoxide (O2-*) generation have both been implicated as important factors associated with the degeneration of serotonergic neurons evoked by methamphetamine (MA) and cerebral ischemia-reperfusion (I-R). Such observations raise the possibility that tryptamine-4,5-dione (T-4,5-D), the major in vitro product of the O2-*-mediated oxidation of 5-HT, might be an endotoxicant that contributes to serotonergic neurodegeneration. When incubated with intact rat brain mitochondria, T-4,5-D (< or = 100 microM) uncouples respiration and inhibits state 3. Experiments with rat brain mitochondrial membrane preparations confirm that T-4,5-D evokes irreversible inhibition of NADH-coenzyme Q1 (CoQ1) reductase and cytochrome c oxidase (COX) apparently by covalently modifying key sulfhydryl (SH) residues at or close to the active sites of these respiratory enzyme complexes. Ascorbic acid blocks the inhibition of NADH-CoQ1 reductase by maintaining T-4,5-D predominantly as 4, 5-dihydroxytryptamine (4,5-DHT), thus preventing its reaction with SH residues. In contrast, ascorbic acid potentiates the irreversible inhibition of COX by T-4,5-D. This may be because the T-4,5-D-4, 5-DHT couple redox cycles in the presence of excess ascorbate and molecular oxygen to cogenerate O2-* and H2O2 that together react with trace levels of iron to form an oxo-iron complex that selectively damages COX. Thus, T-4,5-D might be an endotoxicant that, dependent on intraneuronal conditions, mediates irreversible damage to mitochondrial respiratory enzyme complexes and contributes to the serotonergic neurodegeneration evoked by MA and I-R.


Subject(s)
Endotoxins/metabolism , Indolequinones , Mitochondria/drug effects , Neurodegenerative Diseases/metabolism , Serotonin/metabolism , Superoxides/metabolism , Tryptamines/metabolism , Tryptamines/toxicity , Animals , Ascorbic Acid/pharmacology , Brain Chemistry/drug effects , Electron Transport Complex IV/antagonists & inhibitors , In Vitro Techniques , Male , Oxidation-Reduction , Oxygen Consumption/drug effects , Rats , Rats, Sprague-Dawley
11.
J Neurochem ; 71(5): 2049-62, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9798930

ABSTRACT

We have proposed that a very early step in the pathogenesis of idiopathic Parkinson's disease is the elevated translocation of L-cysteine into neuromelanin-pigmented dopaminergic neurons in the substantia nigra. This influx of L-cysteine was proposed to divert the normal neuromelanin pathway by scavenging dopamine-o-quinone, formed by autoxidation of cytoplasmic dopamine, to give initially 5-S-cysteinyldopamine, which is further oxidized to 7-(2-aminoethyl)-3,4-dihydro-5-hydroxy-2H-1,4-benzothiazine-3-carboxylic acid (DHBT-1). In a recent report, it was demonstrated that DHBT-1 evokes inhibition of complex I respiration when incubated with intact rat brain mitochondria and a time-dependent irreversible inhibition of NADH-coenzyme Q1 (CoQ1) reductase when incubated with mitochondrial membranes. In this study, it is established that the time dependence of NADH-CoQ1 reductase inhibition reflects the oxidation of DHBT-1, catalyzed by an unknown constituent of the inner mitochondrial membrane, to an o-quinone imine intermediate that rearranges to 7-(2-aminoethyl)-5-hydroxy-1,4-benzothiazine-3-carboxylic acid (BT-1) and decarboxylates to 7-(2-aminoethyl)-5-hydroxy-1,4-benzothiazine (BT-2), which are further catalytically oxidized to o-quinone imine intermediates. The electrophilic o-quinone imine intermediates formed in these mitochondria-catalyzed oxidations of DHBT-1, BT-1, and BT-2 are proposed to bind covalently to key sulfhydryl residues at the complex I site, thus evoking irreversible inhibition of NADH-CoQ1 reductase. Evidence for this mechanism derives from the fact that greater than equimolar concentrations of glutathione completely block inhibition of NADH-CoQ1 reductase by DHBT-1, BT-1, and BT-2 by scavenging their electrophilic o-quinone imine metabolites to form glutathionyl conjugates. The results of this investigation may provide insights into the irreversible loss of glutathione and decreased mitochondrial complex I activity, which are both anatomically specific to the substantia nigra and exclusive to Parkinson's disease.


Subject(s)
Brain/metabolism , Enzyme Inhibitors/metabolism , Free Radical Scavengers/pharmacology , Glutathione/antagonists & inhibitors , Mitochondria/metabolism , NAD(P)H Dehydrogenase (Quinone)/antagonists & inhibitors , Thiazines/metabolism , Animals , Brain/drug effects , Catalysis , Dopamine/analogs & derivatives , Dopamine/pharmacology , Electron Transport Complex I , Glutathione/physiology , Male , Mitochondria/drug effects , NADH, NADPH Oxidoreductases/antagonists & inhibitors , Oxidation-Reduction , Parkinson Disease/etiology , Rats , Rats, Sprague-Dawley , Thiazines/pharmacology
12.
Chem Res Toxicol ; 11(7): 824-37, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9671546

ABSTRACT

Iron(II/III) and manganese(II) both catalyze the autoxidation of the neurotransmitter dopamine (DA) in the presence of L-cysteine (CySH) in buffered aqueous solution at pH 7.4. Fe2+/Fe3+ and CySH together generate the hydroxyl (HO.) and cysteinyl thiyl (CyS.) radicals. DA is oxidized by HO. to DA semiquinone radical species that either react with CyS. to give 5-S-cysteinyldopamine (5-S-CyS-DA), 2-S-CyS-DA, and 6-S-CyS-DA or disproportionate to DA-o-quinone that reacts with CySH to give the same cysteinyl conjugates of DA. The major product of this initial reaction is 5-S-CyS-DA. However, 5-S-CyS-DA can be further oxidized by HO. to an o-quinone (2) that undergoes intramolecular cyclization to an o-quinone imine (3). The latter intermediate is the precursor of the dihydrobenzothiazine (DHBT) 7-(2-aminoethyl)-3,4-dihydro-5-hydroxy-2H-1, 4-benzothiazine-3-carboxylic acid (DHBT-1) and several other cyclized products. However, cysteinyl conjugates of DA can also be oxidized by HO. in a one-electron abstraction reaction that leads to DA thiyl radicals. Reactions of these radicals with CyS. or DA semiquinone radicals lead to some novel DA disulfides and thioethers, respectively. The Mn(II)-catalyzed oxidation of DA generates DA-o-quinone that is scavenged by CySH to give 5-S-CyS-DA (major initial product) with lower yields of other cysteinyldopamines. Subsequent Mn(II)-catalyzed oxidation of 5-S-CyS-DA gives o-quinone 2 and thence o-quinone imine 3 that serve as the precursors of DHBT-1 and several other DHBTs. Organic or oxygen radicals do not play significant roles in the Mn(II)-catalyzed oxidation of DA in the presence of CySH. Recent studies have demonstrated that DHBT-1 can be accumulated by brain mitochondria and evoke irreversible inhibition of NADH-coenzyme Q reductase (complex I). Furthermore, iron, manganese, and alterations in glutathione and CySH metabolism have been implicated in the selective degeneration of nigrostriatal dopaminergic neurons in idiopathic and chemically induced Parkinson's disease (PD). Because DHBT-1 is formed in both the iron- and manganese-catalyzed oxidation of DA in the presence of CySH and a defect in mitochondrial complex I respiration contributes to dopaminergic neuronal cell death in PD, the results of this investigation are discussed in terms of their possible implications to an understanding of the neuropathological processes in idiopathic and chemically induced parkinsonism.


Subject(s)
Cysteine/chemistry , Dopamine/chemistry , Dopamine/physiology , Iron/chemistry , Manganese/chemistry , Parkinson Disease/etiology , Chromatography, High Pressure Liquid , Electrochemistry , Magnetic Resonance Spectroscopy , Oxidation-Reduction , Parkinson Disease/physiopathology , Spectrophotometry, Ultraviolet
13.
Chem Res Toxicol ; 11(6): 639-50, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9625732

ABSTRACT

Many new lines of evidence implicate both superoxide anion radical (O2*-) and biogenic amine neurotransmitters in the pathological mechanisms that underlie neuronal damage caused by methamphetamine (MA), glutamate-mediated oxidative toxicity, ischemia-reperfusion, and other neurodegenerative brain disorders. In this investigation the oxidation of 5-hydroxytryptamine (5-HT, serotonin) by an O2*--generating system (xanthine/xanthine oxidase) in buffered aqueous solution at pH 7.4 has been studied. The major product of the O2*--mediated oxidation of 5-HT is tryptamine-4,5-dione (T-4, 5-D). However, O2*- and H2O2, cogenerated by the xanthine oxidase-mediated oxidation of xanthine to uric acid, together react with trace levels of iron that contaminate buffer constituents to give a chemically ill-defined oxo-iron species. This species mediates the oxidation of 5-HT to a C(4)-centered carbocation intermediate that reacts with 5-HT to give 5,5'-dihydroxy-4, 4'-bitryptamine (4,4'-D) and with uric acid to give 9-[3-(2-aminoethyl)-5-hydroxy-1H-indol-4-yl]-2,6,8-triketo-1H,3H, 7H-purine (7) as the major products. These products differ from those formed in the HO*-mediated oxidation of 5-HT under similar conditions. When the reaction is carried out in the presence of the intraneuronal nucleophile glutathione (GSH), T-4,5-D is scavenged to give 7-(S-glutathionyl)tryptamine-4,5-dione, whereas the putative carbocation intermediate is scavenged to give 4-(S-glutathionyl)-5-hydroxytryptamine. T-4,5-D also reacts with the sulfhydryl residues of a model protein, alcohol dehydrogenase, and inhibits its activity. Previous investigators have proposed that T-4, 5-D is a serotonergic neurotoxin. This raises the possibility that T-4,5-D and perhaps other putative intraneuronal metabolites formed by the O2*-/H2O2/oxo-iron-mediated oxidations of 5-HT might be endotoxins that contribute to neurodegeneration in brain regions innervated by serotonergic neurons caused by MA, ischemia-reperfusion, and other neurodegenerative brain disorders.


Subject(s)
Brain Diseases/etiology , Indolequinones , Neurodegenerative Diseases/etiology , Serotonin/metabolism , Superoxides/metabolism , Antioxidants/pharmacology , Free Radical Scavengers/pharmacology , Free Radicals , Glutathione/pharmacology , Humans , Iron Chelating Agents/pharmacology , Oxidation-Reduction , Protein Binding , Tryptamines/metabolism
14.
J Neurochem ; 69(4): 1530-41, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9326282

ABSTRACT

Based on a number of lines of evidence, we have proposed recently that a very early step in the pathogenesis of idiopathic Parkinson's disease might be elevated translocation of L-cysteine into neuromelanin-pigmented dopaminergic cell bodies in the substantia nigra. In vitro studies suggest that such an influx of L-cysteine would divert the neuromelanin pathway by scavenging dopamine-o-quinone, the proximate autoxidation product of dopamine, to give 5-S-cysteinyldopamine, which is oxidized further to 7-(2-aminoethyl)-3,4-dihydro-5-hydroxy-2H-1,4-benzothiazine-3-carboxylic acid (DHBT-1) and other cysteinyldopamines and dihydrobenzothiazines. In this study, it is demonstrated that DHBT-1 inhibits ADP-stimulated oxidation of malate and pyruvate (state 3 or complex I respiration) when incubated with intact rat brain mitochondria with an IC50 of approximatelly 0.80 mM. Incubation of DHBT-1 with freeze-thawed rat brain mitochondria in both the presence and absence of KCN and/or NADH causes an irreversible, time-dependent decrease of NADH-coenzyme Q1 reductase activity. Significantly lower concentrations of DHBT-1 are necessary to cause this effect when mitochondrial membranes are incubated in the absence of KCN and NADH. The irreversible inhibition of mitochondrial complex I caused by DHBT-1 under the latter conditions could be blocked only partially by glutathione, ascorbic acid, superoxide dismutase, or catalase. Together, these results suggest that DHBT-1 can cross the outer mitochondrial membrane and irreversibly inhibit complex I by a mechanism that is not primarily related to oxygen radical-mediated damage. Formation of DHBT-1 requires only dopamine, L-cysteine, and an oxidizing environment, conditions that may well exist in the cytoplasm of neuromelanin-pigmented dopaminergic neurons in the parkinsonian substantia nigra. The results of this study raise the possibility that DHBT-1 might be an endotoxin formed specifically in pigmented dopaminergic neurons that can contribute to irreversible damage to mitochondrial complex I and substantia nigra cell death in Parkinson's disease.


Subject(s)
Mitochondria/metabolism , NAD(P)H Dehydrogenase (Quinone)/antagonists & inhibitors , Thiazines/pharmacology , Animals , Electron Transport Complex I , Endotoxins/metabolism , Enzyme Inhibitors/pharmacology , Male , Mitochondria/physiology , NADH, NADPH Oxidoreductases/antagonists & inhibitors , NADH, NADPH Oxidoreductases/metabolism , Oxygen Consumption/drug effects , Parkinson Disease/metabolism , Rats , Rats, Sprague-Dawley , Substantia Nigra/metabolism
15.
J Neurochem ; 68(5): 1929-41, 1997 May.
Article in English | MEDLINE | ID: mdl-9109519

ABSTRACT

Oxygen radicals have been implicated in the neurodegenerative and other neurobiological effects evoked by methamphetamine (MA) in the brain. It has been reported that shortly after a single large subcutaneous dose of MA to the rat, the serotonergic neurotoxin 5,6-dihydroxytryptamine (5,6-DHT) is formed in the cortex and hippocampus. This somewhat controversial finding suggests that MA potentiates formation of the hydroxyl radical (HO.) that oxidizes 5-hydroxytryptamine (5-HT) to 5,6-DHT, which, in turn, mediates the degeneration of serotonergic terminals. A major and more stable product of the in vitro HO.-mediated oxidation of 5-HT is 5-hydroxy-3-ethylamino-2-oxindole (5-HEO). In this investigation, a method based on HPLC with electrochemical detection (HPLC-EC) has been developed that permits measurement of very low levels of 5-HEO in rat brain tissue in the presence of biogenic amine neurotransmitters/metabolites. After intracerebroventricular administration into rat brain, 5-HEO is transformed into a single major, but unknown, metabolite that can be detected by HPLC-EC. One hour after administration of MA (100 mg/kg s.c.) to the rat, massive decrements of 5-HT were observed in all regions of the brain examined (cortex, hippocampus, medulla and pons, midbrain, and striatum). However, 5-HEO, its unidentified metabolite, or 5,6-DHT were not detected as in vivo metabolites of 5-HT. MA administration, in particular to rats pretreated with pargyline, resulted in the formation of low levels of N-acetyl-5-hydroxytryptamine (NAc-5-HT) in all brain regions examined. These results suggest that MA does not potentiate the HO.-mediated oxidation of 5-HT. Furthermore, the rapid MA-induced decrease of 5-HT might not only be related to oxidative deactivation of tryptophan hydroxylase, as demonstrated by other investigators, but also to the inhibition of tetrahydrobiopterin biosynthesis by NAc-5-HT. The massive decrements of 5-HT evoked by MA are accompanied by small or no corresponding increases in 5-hydroxyindole-3-acetic acid (5-HIAA) levels. This is due, in part, to the relatively rapid clearance of 5-HIAA from the brain and monoamine oxidase (MAO) inhibition by MA. However, the loss of 5-HT without corresponding increases in its metabolites point to other mechanisms that might deplete the neurotransmitter, such as oxidation by superoxide radical anion (O2.-), a reaction that in vitro does not generate 5-HEO or 5,6-DHT but rather another putative neurotoxin, tryptamine-4,5-dione. One hour after administration, MA evokes large depletions of norepinephrine (NE) throughout the brain but somewhat smaller decrements of dopamine (DA) that are restricted to the nigrostriatal pathway. Furthermore, MA evokes a major shift in the metabolism of both NE and DA from the pathway mediated by MAO to that mediated by catechol-O-methyltransferase. The profound and widespread effects of MA on the noradrenergic system, but more anatomically localized influence on the dopaminergic system, suggests that NE in addition to DA, or unusual metabolites of these neurotransmitters, might play roles in the neurodegenerative effects evoked by this drug.


Subject(s)
Brain/drug effects , Brain/metabolism , Dopamine Agents/pharmacology , Methamphetamine/pharmacology , Animals , Dopamine Agents/administration & dosage , Dose-Response Relationship, Drug , Hydroxyl Radical/metabolism , Indoles/metabolism , Injections, Intraventricular , Male , Methamphetamine/administration & dosage , Oxidation-Reduction/drug effects , Oxindoles , Rats , Rats, Sprague-Dawley , Serotonin/analogs & derivatives , Serotonin/metabolism , Serotonin/pharmacokinetics , Time Factors
16.
Alcohol ; 14(3): 213-23, 1997.
Article in English | MEDLINE | ID: mdl-9160798

ABSTRACT

Ethanol is metabolized in the brain by catalase/H2O2 to yield acetaldehyde and by an ethanol-inducible form of cytochrome P450 (P450 IIE1) in a reaction that yields oxygen radicals. Within the cytoplasm of serotonergic axon terminals these metabolic pathways together provide conditions for the endogenous synthesis of 1-methyl-6-hydroxy-1,2,3,4-tetrahydro-beta-carboline (1), by reaction of acetaldehyde with unbound 5-hydroxytryptamine (5-HT), and for the oxygen radical-mediated oxidation of this alkaloid. The major initial product of the hydroxyl radical (HO.)-mediated oxidation of 1 in the presence of free glutathione (GSH), a constituent of nerve terminals and axons, is 8-S-glutathionyl-1-methyl-1,2,3,4-tetrahydro-beta-carboline-5,6-dione (6). When administered into the brains of mice, 6 is a potent toxin (LD50 = 2.9 microg) and evokes episodes of hyperactivity and tremor. Compound 6 binds at the GABA(B) receptor and evokes elevated release and turnover of several neurotransmitters. Furthermore, the GABA(B) receptor antagonist phaclofen attenuates the behavioral response caused by intracerebral administration of 6. These observations suggest that 6 might be an inverse agonist at the GABA(B) receptor site. Accordingly, it is speculated that ethanol drinking might potentiate formation of 6 that contributes to elevated release of several neurotransmitters including dopamine (DA) and endogenous opioids in regions of the brain innervated by serotonergic axon terminals. Subsequent interactions of DA and opioids with their receptors might be related to the initial development of dependence on ethanol. Redox cycling of 6 (and of several putative secondary metabolites) in the presence of intraneuronal antioxidants and molecular oxygen to produce elevated fluxes of cytotoxic reduced oxygen species might contribute to the degeneration of serotonergic pathways. Low levels of 5-HT in certain brain regions of the rat predisposes these animals to drink or augments drinking. Accordingly, 6, formed as a result of ethanol metabolism in the cytoplasm of certain serotonergic axon terminals, might contribute to the initial development of dependence on ethanol, by mediating DA and opioid release, and long-term preference and addiction to the fluid as a result of the progressive degeneration of these neurons.


Subject(s)
Alcoholism/complications , Carbolines/metabolism , Ethanol/metabolism , Substance-Related Disorders/etiology , Animals , Baclofen/analogs & derivatives , Baclofen/pharmacology , Behavior, Animal/drug effects , Brain Diseases/chemically induced , Hydroxyl Radical , Male , Mice , Mice, Inbred ICR , Neurotransmitter Agents/metabolism , Rats , Rats, Sprague-Dawley , Receptors, GABA-B/drug effects
17.
Chem Res Toxicol ; 10(2): 147-55, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9049425

ABSTRACT

Previous studies demonstrated that oxidation of dopamine (DA) in the presence of L-cysteine (CySH) at pH 7.4 gives a complex mixture of cysteinyl conjugates of the neurotransmitter that can be easily further oxidized to a number of dihydrobenzothiazines (DHBTs) along with unidentified yellow products. In this investigation, three of these products have been identified. 7-(2 Aimoethyl)-5-hydroxy-1,4-benzothiazine-3-carboxylic acid (BT-1) is formed as a result of oxidation of 5-S-cysteinyldopamine (5-S-CyS-DA) and 7-(2-aminoethyl)-3,4-dihydro-5-hydroxy-2H-1,4-benzothiazine-3-carboxylic acid (DHBT-1). Regioisomers 6-(2-aminoethyl)-1,8,9,10-tetrahydrobenzo[1,2-b:4,3-b']bis[1,4] thiazine-9-carboxylic acid (12) and 6-(2-aminoethyl)-1,2,3,10-tetrahydrobenzo[1,2-b:4,3-b']bis[1,4] thiazine-2-carboxylic acid (13) are formed by oxidation of 2,5-bi-S-CyS-DA), 6-S-cysteinyl-7-(2-aminoethyl)-3-4-dihydro-5-hydroxy-2H-1, 4-benzothiazine-3-carboxylic acid (DHBT-2) and 6-S-cysteinyl-8(2-aminoethyl)-3,4-dihydro-2H-1,4-benzothiazine-3-carboxy lic acid (DHBT-6). 2,5-Bi-S-CyS-DA, DHBT-2, and DHBT-6 are major early products of DA oxidation in the presence of CySH. However, because these three compounds are the most easily oxidized products formed in this reaction, they are subsequently transformed into 12 and 13, the latter regioisomer always being the major product. Both 12 (LD50 = 18.5 micrograms) and 13 (LD50 = 1.5 microgram) are lethal when administered into the brains of mice and evoke hyperactivity and tremor. The potential relevance of the in vitro chemistry described in this and earlier reports to reaction that might occur in neuromelanin-pigmented dopaminergic neurons in Parkinson's disease is discussed.


Subject(s)
Cysteine/pharmacology , Dopamine/metabolism , Dopamine/toxicity , Animals , Behavior, Animal/drug effects , Brain/drug effects , Dopamine/chemistry , Lethal Dose 50 , Male , Mice , Mice, Inbred ICR , Oxidation-Reduction/drug effects
18.
NIDA Res Monogr ; 173: 146-74, 1997.
Article in English | MEDLINE | ID: mdl-9260188

ABSTRACT

In the event that methamphetamine evokes HO. formation within serotonergic axon terminals, the resultant oxidation of 5-HT would be expected to generate not only 5,6-DHT but also T-4,5-D, 7-S-Glu-T-4,5-D, 6, 8, and 7,7'-D (figure 1), at least three of which (T-4,5-D, 7-S-Glu-T-4,5-D, and 6) are lethal in mouse brain. Furthermore, several intermediates/products formed in the in vitro oxidation of 5-HT by HO. are readily autoxidized (4,5-DHT, 5,6-DHT, 5, 7, and 9) or redox cycled (T-4,5-D, 6, 8, 7,7'-D, 7-S-Glu-T-4,5-D) in reactions that would be expected to yield O2-. and/or H2O2 as byproducts. These byproducts, in the presence of trace levels of transition metal ion catalysts, would be readily converted into HO. (Walling 1975; Halliwell and Gutteridge 1984). Together these putative aberrant oxidative metabolites of 5-HT and HO.-forming reactions might contribute to the degeneration of serotonergic nerve terminals. Similarly, the methamphetamine-induced intraneuronal formation of HO. in dopaminergic terminals might be expected to generate not only 6-OHDA (and 2-OHDA and 5-OHDA, figure 3) but also 5,-S-CyS-DA and 5-S-Glu-DA, precursors of DHBT 17 and other more complex dihydrobenzothiazines (figure 4). DHBTs 17 to 19 are lethal in mouse brain, although at this time the biochemical/chemical mechanisms underlying this toxicity and specific neuronal systems affected are unknown. However, 5-S-CyS-DA and 17 to 19 are much more easily oxidized than DA, and the latter DHBTs appear to be capable of redox cycling reactions (Zhang and Dryhurst 1994). Thus, the HO.-mediated oxidation of DA in dopaminergic nerve terminals induced by methamphetamine might be expected to generate aberrant oxidative metabolites that (as a result of autoxidation and redox cycling reactions) potentiate formation of O2-. and/or H2O2, and then HO. and neuronal damage. A number of lines of evidence, discussed previously, suggest that aberrant metabolite(s) of DA (other than or in addition to 6-OHDA) might contribute to the methamphetamine-induced degeneration of not only dopaminergic terminals but also serotonergic terminals. Similarly, aberrant metabolite(s) of 5-HT (other than or in addition to 5,6-DHT) might be involved in the degeneration of serotonergic and dopaminergic terminals and a subpopulation of cell bodies in the somatosensory cortex. Experimental evidence indicates that some of the neurodegenerative effects evoked by methamphetamine are mediated by NMDA and GABA receptors. Thus, it will be of considerable interest to investigate the neurotoxicity of putative aberrant oxidative metabolites of 5-HT (figures 1 and 2) and DA (figures 4 and 5) towards serotonergic, dopaminergic, and other neuronal systems and their interactions with NMDA, GABA, and other brain receptors. A central question relates to mechanisms by which methamphetamine might evoke the intraneuronal formation of oxygen radicals that appear to play important roles in the overall neurodegenerative processes evoked by this drug (DeVito and Wagner 1989; Cadet et al. 1994). Once putative oxidative metabolites of 5-HT such as T-4,5-D, 7-S-Glu-T-4,5-D, 5,6-DHT, 6, 8, and 7,7'-D (figure 1) are formed intraneuronally, autoxidation/redox cycling reactions should, in principle, be capable of generating O2-. and/or H2O2, the precursors of HO.. Similarly, intraneuronal formation of 6-OHDA, 5-S-CyS-DA, and DHBTs 17 to 19 and 22 would also be expected to potentiate elevated fluxes of O2-., H2O2, and HO. as a result of the facile autoxidation/redox cycling reactions of these putative aberrant metabolites. The presence of very low concentrations of 5-S-CyS-DA in DA-rich regions of human and other mammalian brains suggest that autoxidation (Rosengren et al. 1985; Fornstedt et al. 1986, 1989, 1990) or perhaps some other form of DA oxidation is a normal reaction in vivo. Furthermore, available evidence suggests that it is cytoplasmic DA that is oxidized to give 5-S-CyS-DA (Fornstedt et al. 1989; Fornstedt and


Subject(s)
Brain/drug effects , Methamphetamine/toxicity , Animals , Brain/metabolism , Dopamine/metabolism , Humans , Mice , Oxidation-Reduction , Serotonin/metabolism
19.
Neurosci Lett ; 218(1): 67-71, 1996 Oct 25.
Article in English | MEDLINE | ID: mdl-8939482

ABSTRACT

The present study examined the effects of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and 5,7-dihydroxytryptamine (5,7-DHT) on striatal levels of dopamine (DA), 5-hydroxytryptamine (5-HT), and their metabolites homovanillic acid (HVA) and 5-hydroxyindole-3-acetic acid (5-HIAA), respectively, as well as their influence on locomotor activity in conscious C57BL/6 mice. High doses (s.c., 35-45 mg/kg per day for 10 days) of MPTP resulted in a significant (P < 0.05) increase in locomotor activity and a marked decrease of striatal DA levels. Concomitantly, the ratios of HVA to DA and 5-HIAA to 5-HT increased significantly, the latter reflecting increased 5-HIAA levels. In contrast, i.c.v. administration of the serotonergic neurotoxin 5,7-DHT, either alone or following high doses (40 mg/kg per day for 10 days) of MPTP, decreased locomotor activity. Furthermore, striatal levels of 5-HT and 5-HIAA as well as the 5-HIAA/5-HT ratio decreased significantly. Thus, the increased locomotor activity induced by chronic high doses of MPTP might be due to increased striatal 5-HT levels which appear to compensate for the loss of DA. Furthermore, the locomotor hypoactivity induced by 5,7-DHT may be secondary to the striatal 5-HT deficiency.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , 5,7-Dihydroxytryptamine/pharmacology , Biogenic Amines/metabolism , Corpus Striatum/drug effects , Locomotion/drug effects , Animals , Dose-Response Relationship, Drug , Mice , Mice, Inbred C57BL
20.
Chem Res Toxicol ; 9(7): 1117-26, 1996.
Article in English | MEDLINE | ID: mdl-8902266

ABSTRACT

A very early event in the pathogenesis of idiopathic Parkinson's disease (PD) has been proposed to be an elevated translocation of L-cysteine (CySH) and/or glutathione (GSH) into pigmented dopaminergic cell bodies in the substantia nigra (SN) in which cytoplasmic dopamine (DA) is normally autoxidized to DA-o-quinone as the first step in a reaction leading to black neuromelanin polymer. Such an elevated influx of CySH and GSH would be expected to initially result in formation of 5-S-cysteinyldopamine (5-S-CyS-DA) and 5-S-glutathionyldopamine (5-S-Glu-DA), respectively, and might account for the massive irreversible loss of GSH and progressive depigmentation of SN cells that occurs in the Parkinsonian brain. However, 5-S-Glu-DA has not been detected in the Parkinsonian brain. Furthermore, although the 5-S-CyS-DA/DA and 5-S-CyS-DA/homovanillic acid concentration ratios increase significantly in the SN and cerebrospinal fluid, respectively, of PD patients, the absolute concentrations of 5-S-CyS-DA are extremely low and similar to those measured in age-matched control patients. One explanation for these observations is that 5-S-CyS-DA might be intraneuronally oxidized to more complex cysteinyldopamines and a number of dihydrobenzothiazines (DHBTs) and benzothiazines (BTs). Similarly, 5-S-Glu-DA might be intraneuronally oxidized to more complex glutathionyldopamines. In this investigation, however, it is demonstrated that 5-S-Glu-DA is rapidly metabolized in rat brain to 5-S-CyS-DA and 5-S-(N-acetylcysteinyl) dopamine (5) in reactions mediated by gamma-glutamyl transpeptidase (gamma-GT) and cysteine conjugate N-acetyltransferase. Similarly, 5-S-CyS-DA is metabolized to 5 in rat brain although more slowly than 5-S-Glu-DA. These reactions occur most rapidly in the midbrain, a region that contains the SN. Furthermore, 5, 2-S-(N-acetylcysteinyl)dopamine (6) and 2,5-di-S-(N-acetylcysteinyl)-dopamine (9) are toxic when administered into mouse brain having LD50 values of 14, 25, and 42 micrograms, respectively, and evoke a profound hyperactivity syndrome. These results suggest that the failure to detect 5-S-Glu-DA and the presence of only very low levels of 5-S-CyS-DA in Parkinsonian SN tissue and CSF might be related to both their intraneuronal oxidation and extraneuronal metabolism to N-acetylcysteinyl conjugates of DA. Furthermore, the toxic properties and neurobehavioral responses evoked by 5, 6, and 9 raise the possibility that these N-acetylcysteinyl conjugates of DA, in addition to certain cysteinyldopamines, DHBTs and BTs, might include endotoxins that contribute to SN cell death and other neuronal damage that occurs in PD. Methods are described for the synthesis of several N-acetylcysteinyl conjugates of DA, and their redox behaviors have been studied using cyclic voltammetry.


Subject(s)
Acetylcysteine/metabolism , Acetylcysteine/toxicity , Brain/drug effects , Cysteinyldopa/biosynthesis , Cysteinyldopa/toxicity , Dopamine/metabolism , Dopamine/toxicity , Parkinson Disease/etiology , Animals , Cysteinyldopa/metabolism , Disease Models, Animal , Male , Mice , Mice, Inbred ICR , Oxidation-Reduction , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...