Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
ACS Chem Neurosci ; 10(6): 2977-2988, 2019 06 19.
Article in English | MEDLINE | ID: mdl-30929447

ABSTRACT

Although the exact cause or causes of Parkinson's disease (PD) are not fully understood, it is believed that environmental factors play a major role. The discovery that a synthetic chemical, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-derived N-methyl-4-phenylpyridinium (MPP+), recapitulates major pathophysiological characteristics of PD in humans has provided the strongest support for this possibility. While the mechanism of the selective dopaminergic toxicity of MPP+ has been extensively studied and is, in most respects, well accepted, several key aspects of the mechanism are still debatable. In the present study, we use a series of structurally related, novel, and lipophilic MPP+ derivatives [ N-(2-phenyl-1-propene)-4-phenylpyridinium] to probe the mechanism of action of MPP+ using dopaminergic MN9D and non-neuronal HepG2 cells in vitro. Here we show that effective mitochondrial complex I inhibition is necessary and that the specific uptake through plasma membrane dopamine transporter is not essential for dopaminergic toxicity of MPP+ and related toxins. We also provide strong evidence to support our previous proposal that the selective vulnerability of dopaminergic cells to MPP+ and similar toxins is likely due to the high inherent propensity of these cells to produce excessive reactive oxygen species as a downstream effect of complex I inhibition. Based on the current and previous findings, we propose that MPP+ is the simplest of a larger group of unidentified environmental dopaminergic toxins, a possibility that may have major public health implications.


Subject(s)
1-Methyl-4-phenylpyridinium/toxicity , Dopaminergic Neurons/drug effects , Electron Transport Complex I/drug effects , 1-Methyl-4-phenylpyridinium/analogs & derivatives , Animals , Dopaminergic Neurons/metabolism , Electron Transport Complex I/metabolism , Hep G2 Cells , Humans , Mice , Parkinson Disease/metabolism , Reactive Oxygen Species/metabolism
2.
ACS Chem Neurosci ; 4(5): 858-69, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23647019

ABSTRACT

We have previously introduced fluorescent false neurotransmitters (FFNs) as optical reporters that enable visualization of individual dopaminergic presynaptic terminals and their activity in the brain. In this context, we examined the fluorescent pyridinium dye 4-(4-dimethylamino)phenyl-1-methylpyridinium (APP+), a fluorescent analogue of the dopaminergic neurotoxin MPP+, in acute mouse brain tissue. APP+ is a substrate for the dopamine transporter (DAT), norepinephrine transporter (NET), and serotonin transporter (SERT), and as such represented a candidate for the development of new FFN probes. Here we report that APP+ labels cell bodies of catecholaminergic neurons in the midbrain in a DAT- and NET-dependent manner, as well as fine dopaminergic axonal processes in the dorsal striatum. APP+ destaining from presynaptic terminals in the dorsal striatum was also examined under the conditions inducing depolarization and exocytotic neurotransmitter release. Application of KCl led to a small but significant degree of destaining (approximately 15% compared to control), which stands in contrast to a nearly complete destaining of the new generation FFN agent, FFN102. Electrical stimulation of brain slices at 10 Hz afforded no significant change in the APP+ signal. These results indicate that the majority of the APP+ signal in axonal processes originates from labeled organelles including mitochondria, whereas only a minor component of the APP+ signal represents the releasable synaptic vesicular pool. These results also show that APP+ may serve as a useful probe for identifying catecholaminergic innervations in the brain, although it is a poor candidate for the development of FFNs.


Subject(s)
1-Methyl-4-phenylpyridinium/analogs & derivatives , Adrenergic Neurons/metabolism , Aniline Compounds/metabolism , Brain/metabolism , Dopaminergic Neurons/metabolism , Fluorescent Dyes/metabolism , Pyridinium Compounds/metabolism , Animals , Axons/metabolism , Dopamine Plasma Membrane Transport Proteins/metabolism , Mice , Mitochondria/metabolism , Neostriatum/metabolism , Norepinephrine Plasma Membrane Transport Proteins/metabolism , Presynaptic Terminals/metabolism , Synapses/metabolism , Synaptic Vesicles/metabolism
3.
J Med Chem ; 51(4): 760-8, 2008 Feb 28.
Article in English | MEDLINE | ID: mdl-18220329

ABSTRACT

The active metabolite of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), N-methyl-4-phenylpyridinium (MPP(+)), selectively destroys the dopaminergic neurons and induces the symptoms of Parkinson's disease. Inhibition of mitochondrial complex I and/or the perturbation of dopamine metabolism through cellular and granular accumulation have been proposed as some of the major causes of neurotoxicity. In the present study we have synthesized and characterized a number of MPTP and MPP(+) derivatives that are suitable for the comparative neurotoxicity and complex I inhibition versus dopamine metabolism perturbation studies. Structure-activity studies with bovine chromaffin granule ghosts show that 3'-hydroxy-MPP(+) is one of the best known substrates for the vesicular monoamine transporter (VMAT). A series of compounds that combine the structural features of MPP(+) and a previously characterized VMAT inhibitor, 3-amino-2-phenyl-propene, have been identified as the most effective VMAT inhibitors. These derivatives have been used to define the structural requirements of the VMAT substrate and inhibitor activities.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/analogs & derivatives , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/chemical synthesis , 1-Methyl-4-phenylpyridinium/analogs & derivatives , 1-Methyl-4-phenylpyridinium/chemical synthesis , Vesicular Monoamine Transport Proteins/antagonists & inhibitors , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , 1-Methyl-4-phenylpyridinium/pharmacology , Animals , Cattle , Chromaffin Granules/drug effects , Chromaffin Granules/metabolism , Crystallography, X-Ray , In Vitro Techniques , Models, Molecular , Structure-Activity Relationship , Vesicular Monoamine Transport Proteins/metabolism
4.
J Biol Chem ; 280(19): 19177-84, 2005 May 13.
Article in English | MEDLINE | ID: mdl-15757904

ABSTRACT

The human norepinephrine (NE) transporter (hNET) attenuates neuronal signaling by rapid NE clearance from the synaptic cleft, and NET is a target for cocaine and amphetamines as well as therapeutics for depression, obsessive-compulsive disorder, and post-traumatic stress disorder. In spite of its central importance in the nervous system, little is known about how NET substrates, such as NE, 1-methyl-4-tetrahydropyridinium (MPP+), or amphetamine, interact with NET at the molecular level. Nor do we understand the mechanisms behind the transport rate. Previously we introduced a fluorescent substrate similar to MPP+, which allowed separate and simultaneous binding and transport measurement (Schwartz, J. W., Blakely, R. D., and DeFelice, L. J. (2003) J. Biol. Chem. 278, 9768-9777). Here we use this substrate, 4-(4-(dimethylamino)styrl)-N-methyl-pyridinium (ASP+), in combination with green fluorescent protein-tagged hNETs to measure substrate-transporter stoichiometry and substrate binding kinetics. Calibrated confocal microscopy and fluorescence correlation spectroscopy reveal that hNETs, which are homomultimers, bind one substrate molecule per transporter subunit. Substrate residence at the transporter, obtained from rapid on-off kinetics revealed in fluorescence correlation spectroscopy, is 526 micros. Substrate residence obtained by infinite dilution is 1000 times slower. This novel examination of substrate-transporter kinetics indicates that a single ASP+ molecule binds and unbinds thousands of times before being transported or ultimately dissociated from hNET. Calibrated fluorescent images combined with mass spectroscopy give a transport rate of 0.06 ASP+/hNET-protein/s, thus 36,000 on-off binding events (and 36 actual departures) occur for one transport event. Therefore binding has a low probability of resulting in transport. We interpret these data to mean that inefficient binding could contribute to slow transport rates.


Subject(s)
1-Methyl-4-phenylpyridinium/analogs & derivatives , Symporters/chemistry , 1-Methyl-4-phenylpyridinium/pharmacology , Amphetamine/pharmacology , Biological Transport , Calibration , Cell Line , Cell Membrane/metabolism , Dose-Response Relationship, Drug , Fluorescent Dyes/pharmacology , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/metabolism , Humans , Kinetics , Mass Spectrometry , Microscopy, Confocal , Models, Statistical , Norepinephrine/pharmacology , Norepinephrine Plasma Membrane Transport Proteins , Protein Binding , Pyridinium Compounds/pharmacology , Substrate Specificity , Time Factors
5.
In Vivo ; 18(5): 561-9, 2004.
Article in English | MEDLINE | ID: mdl-15523894

ABSTRACT

1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) is known to induce parkinsonism in humans when it is oxidized to the 1-methyl-4-phenylpyridinium salt (MPP+). We previously reported the syntheses of 1-amino-4-phenyl-1,2,3,6-tetrahydropyridine (APTP) and 1-amino-4-phenyl-pyridinium salt (APP+), the 1-amino analogues of the dopaminergic neurotoxins, MPTP and MPP+, respectively, and demonstrated that both APTP and APP+ are cytotoxic to PC12 cells. In this study, we found that both APTP and APP+ induce apoptotic cell death in PC12 cells. Apoptosis was determined by the Comet assay and flow cytometric analysis. Prior to using the Comet assay for detection of apoptotic PC12 cells, Comet images of apoptotic and necrotic cells were first distinguished by using several standards. Comet images were classified into four groups (A to D) according to their shapes. Class D consisted of the apoptotic cells and was easily distinguished. We also demonstrated that apoptotic and necrotic PC12 cells can be easily differentiated and quantified using the convenient Comet assay.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/analogs & derivatives , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , 1-Methyl-4-phenylpyridinium/pharmacology , Apoptosis/drug effects , DNA Damage , PC12 Cells/drug effects , 1-Methyl-4-phenylpyridinium/analogs & derivatives , Animals , Comet Assay , DNA Fragmentation , DNA, Neoplasm/analysis , DNA, Neoplasm/drug effects , Flow Cytometry , PC12 Cells/pathology , Pyridinium Compounds , Rats
6.
Pharmacol Biochem Behav ; 70(1): 105-14, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11566147

ABSTRACT

1-Methyl-4-phenyl-pyridinium (MPP(+)) and S-adenosyl-L-methionine (SAM) cause Parkinson's disease (PD)-like changes. SAM and MPP(+) require their charged S-methyl and N-methyl groups, so the PD-like symptoms may be related to their ability to modulate the methylation process. The SAM-dependent methylation of phosphatidylethanolamine (PTE) to produce phosphatidylcholine (PTC), via phosphatidylethanolamine-N-methyltransferase (PEMT), and the hydrolysis of PTC to form lyso-PTC, a cytotoxic agent, are potential loci for the action of MPP(+). In this study, the effects of MPP(+) on the methylation of PTE to PTC and the production of lyso-PTC were determined. The results showed that SAM increased PTC and lyso-PTC. The rat striatum showed the highest PEMT activity and lyso-PTC formation, which substantiate with the fact that the striatum is the major structure that is affected in PD. MPP(+) significantly enhanced PEMT activity and the formation of lyso-PTC in the rat liver and brain. MPP(+) increased the affinity and the V(max) of PEMT for SAM. 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) effect was lesser and inhibited by deprenyl (MAO-B inhibitor). The nor-methyl analogs of MPP(+) were inactive, but some of the charged analogs of MPP(+) showed comparable effects to those of MPP(+). Lyso-PTC that can be increased by SAM and MPP(+) caused severe impairments of locomotor activities in rats. These results indicate that SAM and MPP(+) have complementary effects on phospholipid methylation. Thus, SAM-induced hypermethylation could be involved in the etiology of PD and an increase of phospholipid methylation could be one of the mechanisms by which MPP(+) causes parkinsonism.


Subject(s)
1-Methyl-4-phenylpyridinium/pharmacology , Herbicides/pharmacology , Methyltransferases/metabolism , Phospholipids/metabolism , 1-Methyl-4-phenylpyridinium/analogs & derivatives , Animals , Brain/drug effects , Brain/enzymology , Dose-Response Relationship, Drug , Liver/drug effects , Liver/enzymology , Male , Methylation/drug effects , Motor Activity/drug effects , Motor Activity/physiology , Rats , Rats, Sprague-Dawley
7.
Brain Res ; 910(1-2): 116-25, 2001 Aug 10.
Article in English | MEDLINE | ID: mdl-11489261

ABSTRACT

The vesicular monoamine transporter 2 (VMAT2) has sequence homology with bacterial multidrug transporters which in turn share homology with mammalian P-glycoprotein (P-GP). Both VMAT2 and P-GP can detoxify cells. 1-Methyl-4-phenylpyridinium (MPP(+)), the toxic metabolite of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), is a substrate for VMAT2 that has several structural features in common with P-GP substrates and inhibitors. The present studies investigated whether P-GP is responsible for the elimination of MPP(+) from the brain. Additionally, VMAT2 and P-GP are inhibited by many of the same compounds. Thus we also investigated whether VMAT2 inhibitors could block P-GP in vitro and vice versa whether P-GP inhibitors could block VMAT2 mediated transport of [3H]-DA into synaptic vesicles. In mice treated with MPTP and a P-GP inhibitor (quinidine, trans-flupentixol or cyclosporine A), the elimination of MPP(+) from the striatum was significantly delayed. However, in experiments using various cell lines expressing either mouse or human P-GP, MPP(+) did not reverse the P-GP mediated resistance to vincristine, suggesting that MPP(+) is a poor substrate for P-GP. Additional experiments were performed using mdr1a/b double knockout mice which lack functional P-GP encoded by these two genes. Data from mdr1a/b knockout mice treated with MPTP also suggest that MPP(+) is not extruded from the brain by P-GP. In other studies, we demonstrated that the VMAT2 inhibitors tetrabenazine and Ro 4-1284 inhibit P-GP and that the P-GP inhibitors trans-flupentixol and quinidine inhibit VMAT2. Thus, several new drugs can be added to the list of compounds that are able to inhibit both VMAT2 and P-GP, providing further evidence of the similarity between these two transporters.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacokinetics , 1-Methyl-4-phenylpyridinium/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Brain/drug effects , Drug Interactions/physiology , Membrane Glycoproteins/antagonists & inhibitors , Membrane Transport Proteins , Neuropeptides , Parkinsonian Disorders/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/metabolism , 1-Methyl-4-phenylpyridinium/analogs & derivatives , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Anti-Arrhythmia Agents/pharmacology , Brain/metabolism , Brain/physiopathology , Calcium Channel Blockers/pharmacology , Cell Survival/drug effects , Cell Survival/physiology , Dopamine/metabolism , Dopamine Antagonists/pharmacology , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Immunosuppressive Agents/pharmacology , Male , Membrane Glycoproteins/metabolism , Mice , Mice, Knockout , Parkinsonian Disorders/physiopathology , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism , Tumor Cells, Cultured , Vesicular Biogenic Amine Transport Proteins , Vesicular Monoamine Transport Proteins
8.
J Biochem Toxicol ; 11(1): 33-43, 1996.
Article in English | MEDLINE | ID: mdl-8806050

ABSTRACT

Inhibition of NADH dehydrogenase (Complex I) of the mitochondrial respiratory chain by 1-methyl-4-phenylpyridinium (MPP+) and its analogs results in dopaminergic cell death. In the present study, the inhibition of mitochondrial respiration and of NADH oxidation in inverted inner membrane preparations by the oxidation products of N-methyl-stilbazoles (N-methyl-styrylpyridiniums) are characterized. These nonflexible MPP+ analogs were found to be considerably more potent inhibitors than the corresponding MPP+ derivatives. The IC50 values for these compounds and previously published figures for MPP+ analogs were then used to select a computer model based on structural parameters to predict the inhibitory potency of other compounds that react at the "rotenone site" in Complex I. A series of 12 novel inhibitors different in structure from the basic set were used to test the predictive capacity of the models selected. Despite major structural differences between the novel test compounds and the MPP+ and styrylpyridinium analogs on which the models were based, substantial agreement was found between the predicted and experimentally determined IC50 values. The value of this technique lies in the potential for the prediction of the inhibitory potency of other drugs and toxins which block mitochondrial respiration by interacting at the rotenone sites.


Subject(s)
1-Methyl-4-phenylpyridinium/analogs & derivatives , 1-Methyl-4-phenylpyridinium/toxicity , Dopamine Agents/toxicity , Herbicides/toxicity , NADH Dehydrogenase/antagonists & inhibitors , Animals , Cell Membrane/drug effects , Dose-Response Relationship, Drug , Lethal Dose 50 , Mitochondria, Liver/drug effects , Oxidation-Reduction , Oxygen Consumption/drug effects , Pyridinium Compounds/metabolism , Pyridinium Compounds/toxicity , Rats , Structure-Activity Relationship
9.
Neurosci Lett ; 199(3): 199-202, 1995 Oct 27.
Article in English | MEDLINE | ID: mdl-8577397

ABSTRACT

The effects of N-methyl-4-phenylpyridinium (MPP+) and its endogenous analog, 2,9-di-methyl-norharmanium (2,9-Me2NH+), on in vivo tyrosine hydroxylation were evaluated in freely moving rats. MPP+ gradually but almost completely reduced tyrosine hydroxylation, even at a dose as low as 0.05 mM. This effect was considered to be caused by the inhibition of tyrosine hydroxylase (TH) activation. On the contrary, 1 mM 2,9-Me2NH+ rapidly reduced 3,4-dihydroxyphenylalanine production to 10% of the basal level only during its perfusion, indicating direct inhibition of TH activity. The present study revealed that MPP+ and 2,9-Me2NH+ were taken up into dopaminergic neurons and then inhibited in vivo dopamine synthesis prior to cell death possibly in different manners.


Subject(s)
1-Methyl-4-phenylpyridinium/pharmacology , Carbolines/pharmacology , Corpus Striatum/drug effects , Tyrosine 3-Monooxygenase/drug effects , 1-Methyl-4-phenylpyridinium/analogs & derivatives , Analysis of Variance , Animals , Dose-Response Relationship, Drug , Male , Rats , Rats, Wistar
10.
Biochem J ; 306 ( Pt 2): 359-65, 1995 Mar 01.
Article in English | MEDLINE | ID: mdl-7887889

ABSTRACT

N-methyl-4-phenylpyridinium (MPP+), the neurotoxic metabolite of N-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, kills dopaminergic neurons after its accumulation in mitochondria where it inhibits Complex I of the respiratory chain. MPP+ inhibits respiration by binding to both a hydrophobic and a hydrophilic site on Complex I and this inhibition is increased by the lipophilic tetraphenylboron anion (TPB-) which facilitates movement of MPP+ through membranes and its penetration to the hydrophobic binding site on Complex I. To investigate the inhibition of respiration by MPP(+)-like compounds, we have measured simultaneously NADH-linked mitochondrial respiration and the uptake and accumulation of the N-benzyl-4-styrylpyridinium and N-ethyl-4-styrylpyridinium cations in mitochondria using ion-selective electrodes. The data provide direct evidence that TPB- increases the inhibition not by increasing matrix concentration but by facilitating access to the inhibitory sites on Complex I. We have also compared the rates of uptake of MPP+ analogues of varied lipophilicity by the inner membrane and the development of inhibition of NADH oxidation, using an inverted mitochondrial inner membrane preparation and appropriate ion-selective electrodes. These experiments demonstrated that the amount of MPP+ analogue bound to the inner membrane greatly exceeded the quantity required for complete inhibition of NADH oxidation. Moreover, binding to the membrane occurred much more rapidly than the development of inhibition with all MPP+ analogues tested. This suggests that the attainment of a correct orientation of these compounds within the membrane and the binding site may be a rate-limiting step in the development of inhibition.


Subject(s)
1-Methyl-4-phenylpyridinium/analogs & derivatives , Electron Transport , Ion-Selective Electrodes , Mitochondria, Liver/metabolism , NAD(P)H Dehydrogenase (Quinone)/antagonists & inhibitors , 1-Methyl-4-phenylpyridinium/metabolism , 1-Methyl-4-phenylpyridinium/pharmacology , Animals , Cations , Cattle , Female , Intracellular Membranes/metabolism , Mitochondria, Heart , NAD/metabolism , Oxidation-Reduction , Pyridinium Compounds/metabolism , Rats , Rats, Wistar
11.
Brain Res ; 675(1-2): 279-88, 1995 Mar 27.
Article in English | MEDLINE | ID: mdl-7796140

ABSTRACT

Eleven beta-carbolinium compounds (beta C+s) and MPP+ were stereotaxically injected (40-200 nmol in 5 microliter of vehicle) unilaterally into the substantia nigra of anesthetized adult male Sprague-Dawley rats. The rats were sacrificed after three weeks. The ipsilateral striatum was analyzed for dopamine and DOPAC levels with HPLC. The brainstem injection site was fixed and cut coronally. The largest lesion area in each animal was measured using NIH IMAGE. Three beta C+s produced lesions whose mean areas were nearly as large as that produced by MPP+ (defined as 100%): 2,9-Me2-harman (94%), 2-Me-harmol (74%), and 2,9-Me2-norharman (57%). Three other compounds produced somewhat smaller lesions: 2-Me-harmaline (34%), 6-MeO-2-Me-harman (29%), and 2-Me-harmine (25%). The remaining compounds were ineffective (< or = 12%): norharman, 2-Me-norharman, 2-Me-harman, harmine, and 2-Me-6-MeO-harmalan. A 40 nmol dose of MPP+ reduced ipsilateral striatal dopamine to 0.6% of control. None of the beta C+s approached this, although several did significantly reduce striatal dopamine at doses of either 40 nmol (2,9-Me2-harman (37%), 2,9-Me2-norharman (42%), and 2-Me-harman (63%)) or 200 nmol (2-Me-harmaline (23%), norharman (63%), and 2-Me-norharman (64%)). There was a moderate negative correlation between lesion size and dopamine level (r = -0.65). There were also moderately strong correlation between lesion size and dopamine level (r = -0.65). There were also moderately strong correlations (r = 0.39-0.78) between the beta C+ nigral lesion area or striatal dopamine level potencies and their previously described IC50 values for inhibiting mitochondrial respiration or their toxicity to PC12 cells in culture. Interestingly, our correlation analysis revealed a remarkably strong correlation between beta C+ Ki MAO-A values and their toxicity to PC12 LDH release (r = -0.84) or PC12 protein loss (r = 0.79). Although beta C+s appear to be less specific toxins than MPP+, their levels in human substantia nigra are 8-20-fold higher than in cortex, making their role as relatively selective nigral toxins in Parkinson's disease plausible.


Subject(s)
1-Methyl-4-phenylpyridinium/analogs & derivatives , 1-Methyl-4-phenylpyridinium/toxicity , Neostriatum/metabolism , Substantia Nigra/metabolism , 1-Methyl-4-phenylpyridinium/administration & dosage , 3,4-Dihydroxyphenylacetic Acid/metabolism , Animals , Chromatography, High Pressure Liquid , Dopamine/metabolism , Image Processing, Computer-Assisted , Injections , Male , Neostriatum/drug effects , Neostriatum/pathology , Rats , Rats, Sprague-Dawley , Substantia Nigra/drug effects , Substantia Nigra/pathology
12.
J Neurochem ; 63(2): 655-61, 1994 Aug.
Article in English | MEDLINE | ID: mdl-8035189

ABSTRACT

1-Methyl-4-phenylpyridinium (MPP+), the toxic agent in MPTP-induced dopaminergic neurotoxicity, is thought to act by inhibiting mitochondrial electron transport at complex I. This study examined this latter action further with a series of 4'-alkylated analogues of MPP+. These derivatives had IC50 values that ranged from 0.5 to 110 microM and from 1.6 to 3,300 microM in mitochondria and electron transport particles (ETPs), respectively. The IC50 values of corresponding 4'-alkylated phenylpyridine derivatives to inhibit NADH-linked oxidation ranged from 10 to 205 microM in mitochondria and from 1.7 to 142 microM in ETPs. The potencies of both classes of inhibitors directly correlated with their ability to partition between 1-octanol and water. In mitochondria, increased hydrophobicity resulted in greater inhibition of NADH dehydrogenase but a smaller dependence on the transmembrane electrochemical gradient for accumulation of the pyridiniums as evidenced by an approximately 600-fold, versus only a 36-fold, increase in the IC50 of MPP+ versus 4'-pentyl-MPP+, respectively, in the presence of uncoupler. In ETPs, the analogous increase in potencies of the more hydrophobic analogues was also consistent with an inhibitory mechanism that relied on differential partitioning into the lipid environment surrounding NADH dehydrogenase. However, the pyridinium charge must play a major role in explaining the inhibitory mechanism of the pyridiniums because their potencies are much greater than would be predicted based solely on hydrophobicity. For example, in ETPs, 4'-decyl-MPP+ was nearly 80-fold more potent than phenylpyridine although the latter compound partitions twice as much into 1-octanol.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
1-Methyl-4-phenylpyridinium/analogs & derivatives , 1-Methyl-4-phenylpyridinium/pharmacology , Mitochondria, Heart/metabolism , Mitochondria, Liver/metabolism , Neurotoxins/pharmacology , Pyridines/pharmacology , Animals , Cattle , Mice , Mitochondria, Heart/drug effects , Mitochondria, Liver/drug effects , NAD(P)H Dehydrogenase (Quinone)/antagonists & inhibitors , NADH Dehydrogenase/metabolism , Structure-Activity Relationship , Uncoupling Agents/pharmacology
13.
J Neurochem ; 62(4): 1503-10, 1994 Apr.
Article in English | MEDLINE | ID: mdl-8133278

ABSTRACT

N-Methylated beta-carbolinium cations that can form in vivo from environmental or endogenous beta-carbolines are putative neurotoxic factors in Parkinson's disease. The cytotoxicities of 11 N-methylated beta-carbolinium cations and N-methyl-4-phenylpyridinium cation (MPP+), the experimental parkinsonian neurotoxicant which the carbolinium cations structurally resemble, were examined using rat pheochromocytoma (PC12) cells cultured in "low energy" N-5 medium; cell death was estimated by released lactate dehydrogenase activity and viable cell protein. Of the eight N2-monomethylated beta-carbolinium cations utilized, only 2-methyl-harmalinium (harmaline-2-methiodide) was as cytotoxic as MPP+. Also, three N2(beta), N9(indole)-dimethylated beta-carbolinium cations displayed cytotoxic effects, with the simplest, 2,9-dimethylnorharmanium, approaching the effectiveness of MPP+ in PC12 cells cultured in N-5 medium. However, when PC12 cells grown in higher energy Dulbecco's modified Eagle's medium were utilized with selected effective cations, it was observed that the cultures were relatively resistant to MPP+ and 2,9-dimethylnorharmanium, but remained vulnerable to 2-methylharmalinium. The results are interpreted to mean that different cytotoxic mechanisms exist for the two most potent beta-carbolinium cations--namely, a mechanism for the 2,9-dimethyl-beta-carbolinium species that, as with MPP+, is conditional on mitochondrial ATP depletion, but a different (or additional) mechanism for 2-methylharmalinium that is independent of mitochondrial inhibition. The possible accumulation of these cytotoxic cations in Parkinson's disease is discussed in the context of these findings.


Subject(s)
1-Methyl-4-phenylpyridinium/analogs & derivatives , 1-Methyl-4-phenylpyridinium/toxicity , Harmaline/analogs & derivatives , PC12 Cells/drug effects , Animals , Cations , Cell Death/drug effects , Culture Media , Energy Metabolism , Harmaline/chemistry , Harmaline/toxicity , Kinetics , L-Lactate Dehydrogenase/metabolism , Methylation , Molecular Structure , Parkinson Disease/etiology , Rats
14.
J Biol Chem ; 269(5): 3167-74, 1994 Feb 04.
Article in English | MEDLINE | ID: mdl-8106350

ABSTRACT

We have investigated the mechanism of the inhibition of membrane-bound NADH dehydrogenase by 1-methyl-4-phenylpyridinium (MPP+) and a series of its 4'-alkyl-substituted analogs of increasing hydrophobicity, as well as their neutral, desmethyl congeners. Comparison of hydrophobicity, as measured by partition coefficients, with the IC50 for the inhibition of NADH oxidase activity in mitochondrial inner membrane preparations shows a negative correlation, but the cationic inhibitors are more effective than the neutral analogs with similar hydrophobicity. The presence of 10 microM tetraphenylboron (TPB-) potentiates the inhibitory power of positively charged analogs up to 4'-pentyl-MPP+, while the neutral inhibitors are unaffected by TPB-. Without TPB-, the more hydrophilic analogs give incomplete inhibition, but the inclusion of TPB- permits the attainment of complete inhibition, accompanied by the appearance of sigmoidal titration curves. These data support the hypothesis that MPP+ analogs, like rotenone, are bound at two sites on the enzyme and occupancy of both is required for complete inhibition. TPB-, by forming ion pairs with the cationic analogs, facilitates their equilibration to both sites in membrane preparations. When present in molar excess over the MPP+ analog, TPB- partially reverses the inhibition by decreasing its concentration in the more hydrophilic binding site. The effect of temperature and of pH on the IC50 values for inhibition support the concept of dual binding sites, and the pH dependence of the inhibition reveals the participation of two ionized protein groups in the binding, one of which may be a thiol group.


Subject(s)
1-Methyl-4-phenylpyridinium/analogs & derivatives , 1-Methyl-4-phenylpyridinium/pharmacology , Intracellular Membranes/metabolism , Multienzyme Complexes/antagonists & inhibitors , NAD(P)H Dehydrogenase (Quinone)/antagonists & inhibitors , NADH Dehydrogenase/antagonists & inhibitors , NADH, NADPH Oxidoreductases/antagonists & inhibitors , Pyridines/pharmacology , 1-Methyl-4-phenylpyridinium/chemical synthesis , 1-Methyl-4-phenylpyridinium/chemistry , Binding Sites , Dose-Response Relationship, Drug , Electron Transport/drug effects , Intracellular Membranes/drug effects , Kinetics , Mitochondria/enzymology , Pyridines/chemical synthesis , Pyridines/chemistry , Rotenone/metabolism , Structure-Activity Relationship
15.
J Med Chem ; 37(1): 151-7, 1994 Jan 07.
Article in English | MEDLINE | ID: mdl-8289189

ABSTRACT

1-Methyl-1,2,3,6-tetrahydrostilbazole (MTHS) and its analogs are oxidized by monoamine oxidase (MAO) A at slow rates comparable to that for the structurally similar neurotoxin, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, but the rates of oxidation by MAO B vary over a wide range depending on the structure of the analog. MAO A oxidation of all of the analogs yielded nonhyperbolic kinetic patterns, with little difference between the cis and trans isomers. In contrast MAO B showed hyperbolic kinetics and distinct stereoselectivity for the cis isomers. The corresponding pyridinium forms of trans-MTHS and its analogs were more potent inhibitors of MAO A (Ki values between 0.3 and 5 microM) than of MAO B, for which the Ki values varied greatly. The data suggest that the stringency of the MAO A active site for the geometry of the substrate molecule is less strict than that of MAO B. With MAO B, any substitution on the phenyl ring can lead to dramatic changes in the substrate properties which may be explained by the different orientation of substrate at the active site of the enzyme. Molecular geometry but not the effects of the substituents was shown to be an important factor in determining the effectiveness of substrate oxidation by MAO B.


Subject(s)
Isoenzymes/metabolism , Monoamine Oxidase/metabolism , Pyridines/metabolism , Styrenes/metabolism , 1-Methyl-4-phenylpyridinium/analogs & derivatives , Binding Sites , Chemical Phenomena , Chemistry, Physical , Kinetics , Molecular Structure , Monoamine Oxidase Inhibitors/pharmacology , Oxidation-Reduction , Pyridines/chemistry , Pyridines/pharmacology , Stereoisomerism , Structure-Activity Relationship , Styrenes/chemistry , Styrenes/pharmacology , Substrate Specificity
16.
J Neurochem ; 61(4): 1546-8, 1993 Oct.
Article in English | MEDLINE | ID: mdl-8377005

ABSTRACT

Expression of the neurotoxicity of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, following oxidation to 1-methyl-4-phenylpyridinium ion (MPP+), is believed to involve inhibition of mitochondrial electron transport from NADH dehydrogenase (complex I) to ubiquinone. MPP+ and its analogues have been shown to block electron transport at or near the same site as two powerful inhibitors of mitochondrial respiration, rotenone and piericidin A. All three types of inhibitors combine at two sites on NADH dehydrogenase, a hydrophilic and hydrophobic one, and occupancy of both sites is required for complete inhibition. Tetraphenylboron anion (TPB-) in catalytic amounts is known to increase the effectiveness of positively charged MPP+ analogues in blocking mitochondrial respiration. A part of this effect involves facilitation of the entry of MPP+ congeners into the hydrophobic site by ion pairing, as has been demonstrated in studies with submitochondrial particles (electron transport particles). This communication documents the fact that TPB-, when present in molar excess over the MPP+ analogues, reverses the inhibition. This seems to involve again strong ion pairing, removal of the inhibitory analogue from one to the two binding sites, and concentration of the inhibitor in the membrane, so that only the hydrophobic binding site remains occupied, resulting in lowering of the inhibition to 30-40%.


Subject(s)
1-Methyl-4-phenylpyridinium/analogs & derivatives , NADH Dehydrogenase/antagonists & inhibitors , Animals , Cattle , Enzyme Activation/drug effects , NADH Dehydrogenase/metabolism , Tetraphenylborate/pharmacology
17.
Arch Biochem Biophys ; 296(2): 482-8, 1992 Aug 01.
Article in English | MEDLINE | ID: mdl-1321585

ABSTRACT

Several derivatives of 1-methyl-4-phenylpyridinium (MPP+), i.e., 1-methyl-4-(4'-nitrophenyl)pyridinium (1), 1-methyl-4-(4'-cyanophenyl)pyridinium (2), 1-methyl-4-(3'-nitrophenyl)pyridinium (3), 1-methyl-4-(4'-chlorophenyl)pyridinium (4), 1-methyl-4-(4'-acetamidophenyl)pyridinium (5), and 1-methyl-4-(4'-aminophenyl)pyridinium (6), were synthesized in order to compare their toxicity with that of paraquat (PQ2+) in Escherichia coli. Addition of compounds 1, 2, and 3 to aerobic E. coli cell suspensions caused extracellular ferricytochrome c reduction, which was inhibited by superoxide dismutase in the same manner as that in the case of PQ2+. The rate of the ferricytochrome c (cyt. c) reduction was in the order of PQ2+ greater than 1 greater than 2 greater than 3, which is the same as that of the redox potentials of these compounds. On the other hand, MPP+, 4, 5, and 6, which have more negative potentials, had no effect on the cyt. c reduction. Compound 1 inhibited the growth of E. coli under aerobic conditions, but not under anaerobic conditions. The results show that compound 1 can act as a mediator for production of superoxide (O2-.), which seriously injures E. coli cells. However, though compounds 2 and 3 catalyzed the production of O2-. in E. coli cells, their activity of O2-. production was much lower than that of compound 1 or PQ2+. Thus, compound 3 had no effect on growth or survival of E. coli at 1 mM, while compounds 2 and 4 had both bacteriostatic and bacteriocidal effects which were independent of dioxygen (O2). The results show that the toxic mechanism is different from that of compound 1. MPP+, 5, and 6 had no effect on growth of E. coli. This paper shows that compound 1 is a novel enhancer of intracellular superoxide production, though the mechanism of toxicity of compounds 2 and 4 is not clear yet. The results suggest that the redox potential is a crucial factor for manifestation of the activity.


Subject(s)
1-Methyl-4-phenylpyridinium/analogs & derivatives , Escherichia coli/drug effects , Paraquat/pharmacology , 1-Methyl-4-phenylpyridinium/pharmacology , Cytochrome c Group/metabolism , Escherichia coli/growth & development , Escherichia coli/metabolism , Kinetics , Molecular Structure , Oxidation-Reduction , Superoxides/metabolism
20.
J Pharmacol Exp Ther ; 260(3): 1400-9, 1992 Mar.
Article in English | MEDLINE | ID: mdl-1312170

ABSTRACT

Several analogs of 1-methyl-4-phenylpyridinium (MPP+) were evaluated for their affinity for the dopamine uptake system and their ability to inhibit NADH dehydrogenase (complex I) of the mitochondrial electron-transport chain. Moreover, these compounds were tested for their ability to cause selective dopaminergic neurotoxicity in cultured mesencephalic neurons. Simultaneous [3H]dopamine and gamma-amino-[14C]butyric acid uptake and immunocytochemical techniques were used as indices of neuronal damage in cultured cells. The compounds that were potent and selective dopaminergic neurotoxins had high affinity for the dopamine transport system, as measured by their ability to cause dopamine release, and were similar to MPP+ in inhibiting mitochondrial respiration. One compound (1-methyl-4-phenylpyrimidinium) had high affinity for the dopamine uptake system but was a weak inhibitor of mitochondrial respiration and, accordingly, was not neurotoxic. The 4'-alkylated analogs of MPP+, which were poor substrates for the dopamine uptake system and extremely potent inhibitors of mitochondrial respiration, caused a nonselective damage of neurons in culture. Analogs that were not substrates for the dopamine carrier and not inhibitors of mitochondrial respiration were not neurotoxic. This study describes the neurotoxicity of a number of analogs of MPP+ and highlights the importance of the dopamine uptake system and the ability to inhibit mitochondrial respiration as critical processes in conferring selectivity and neurotoxicity, respectively, to MPP+ and analogs, for dopaminergic neurons in culture.


Subject(s)
1-Methyl-4-phenylpyridinium/toxicity , Dopamine/metabolism , Mitochondria/drug effects , Neurons/drug effects , Oxygen Consumption/drug effects , Receptors, Dopamine/drug effects , 1-Methyl-4-phenylpyridinium/analogs & derivatives , Animals , Cells, Cultured , Lethal Dose 50 , Mitochondria/metabolism , Rats , Receptors, GABA-A/drug effects , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...