Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Gene Ther ; 8(1): 41-8, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11402300

ABSTRACT

Adenovirus-mediated gene therapy of bladder diseases has been limited by the inability to transduce the urothelium successfully using adenoviral vectors. We have sought to identify agents that would increase adenovirus-mediated transgene expression in the bladder. We have utilized a rat model to screen compounds for their ability to enhance viral transgene expression in the rat bladder. Rats received intravesical administration of replication-deficient adenovirus (rAd) formulated in various agents, and transgene expression was evaluated after 48 h by determining the amount of lacZ expression in the luminal epithelium of the bladder. We report the identification of two different polyamides, each capable of dramatically increasing viral transgene expression in the bladder without causing detectable alteration of the umbrella cell layer of the urothelium. We have utilized a carcinogen-induced rat bladder tumor model to demonstrate that these polyamides are also capable of enhancing viral transgene expression in tumor tissue. The identification of these polyamides potentiates the use of adenovirus-mediated gene therapy for the treatment of superficial bladder cancer or other bladder diseases.


Subject(s)
Gene Expression Regulation, Viral/drug effects , Genetic Therapy/methods , Nylons/pharmacology , Urinary Bladder Neoplasms/therapy , Urothelium/metabolism , Adenoviridae/genetics , Animals , Female , Genetic Vectors , Male , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley , Transgenes/genetics , Urinary Bladder Neoplasms/pathology , Urothelium/ultrastructure
2.
J Interferon Cytokine Res ; 21(12): 1103-15, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11798469

ABSTRACT

The type I interferon-alpha (IFN-alpha) family is a family of natural small proteins that have clinically important anti-infective and antitumor activity. We have developed a semisynthetic protein-polymer conjugate of IFN-alpha2b (Intron A) by attaching a 12,000-Da monomethoxypolyethylene glycol (PEG-12000) polymer to the protein. PEG conjugation is thought to increase the serum half-life and thereby prolong patient exposure to IFN-alpha2b without altering the biologic potency to the protein. Matrix-assisted laser desorption ionization/mass spectrometry (MALDI-MS), high-performance size exclusion chromatography (HPSEC), circular dichroism (CD) analysis and tryptic digestion peptide analysis of PEG Intron demonstrated that the IFN-alpha2b protein was approximately 95% monopegylated and that the primary, the secondary, and the tertiary structures were unaltered. Pegylation did not affect the epitope recognition of antibodies used for Intron A quantitation. An extensive analysis of the pegylated positional isomers revealed that approximately 50% of PEG Intron was monopegylated on the His(34) residue of the IFN-alpha2b protein. The highest antiviral activity of the pegylated positional isomers for PEG Intron was associated with the His(34) pegylated isomer. The specific activity for PEG Intron in an antiviral cytopathic protection assay was 28%, relative to Intron A. However, the potency of PEG Intron, defined as bioactivity independent of protein concentration, was comparable to Intron A at both the molecular and cellular levels in a battery of in vitro assays. Equivalent units of PEG Intron and Intron A were indistinguishable for the induction of several key IFN-induced genes, including 2',5'-oligoadenylate synthetase (2',5'-OAS) and protein kinase R (PKR), in Molt 4 cells. The antiviral dose-response curves revealed that there were no significant differences between PEG Intron and Intron A. This demonstrated that the introduction of more IFN-alpha2b protein associated with equivalent unit dosing of PEG Intron did not create any antagonism or agonism in the antiviral assay. In assays for the immune response, PEG Intron and Intron A displayed comparable potency for both natural-killer (NK) and lymphokine-activated killer (LAK) cell cytolytic activity and for the induction of class I major histocompatibility protein. These results demonstrate that PEG Intron maintains an in vitro biologic potency profile for both antiviral and immunotherapeutic activity that is highly comparable to that of Intron A.


Subject(s)
Antineoplastic Agents/chemistry , Antiviral Agents/chemistry , Interferon-alpha/chemistry , Polyethylene Glycols/chemistry , Antineoplastic Agents/pharmacology , Antiviral Agents/pharmacology , Cells, Cultured , Chromatography, Gel , Circular Dichroism , Cytotoxicity Tests, Immunologic , Dose-Response Relationship, Drug , Humans , Interferon alpha-2 , Interferon-alpha/pharmacology , Isomerism , Models, Molecular , Polyethylene Glycols/pharmacology , RNA, Messenger/analysis , Recombinant Proteins , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Structure-Activity Relationship , Tumor Cells, Cultured
3.
Biochemistry ; 39(35): 10634-40, 2000 Sep 05.
Article in English | MEDLINE | ID: mdl-10978146

ABSTRACT

Interferons display a wide range of antiviral, antiproliferative, and immunomodulatory activities on a variety of cell types and have been used to treat many diseases including hairy-cell leukemia and hepatitis B and C and have also been applied to other therapeutic areas. To improve the pharmacological properties of interferon (IFN) alpha-2b, a long-acting pegylated form (PEG-IFN) has been developed [PEG, monomethoxy poly(ethylene glycol) with average molecular mass of 12 000 Da]. PEG-IFN is a mixture of pegylated proteins with differing sites of PEG attachment. To identify the major positional isomer in the pegylated material [PEG-IFN(His-34)], NMR studies were conducted on a subtilisin-digested N-acetylated peptide of the major positional isomer [PEG-IFN(His-34)dig], synthetic peptide analogues containing His-34, as well as unmodified IFN and PEG-IFN(His-34). Our studies reveal a novel interferon-polymer attachment site as a histidine-linked interferon conjugate. We show that the major component of PEG-IFN is pegylated in the imidazole side chain of histidine-34. Chemical shift data suggest that pegylation occurs mainly at the N(delta)(1) position in the imidazole side chain of this residue. This positional isomer, PEG-IFN(His-34), comprises approximately 47% of the total pegylated species when PEG-IFN is synthesized under the current experimental conditions at pH 6.5 with an electrophilic derivative of PEG, succinimidyl carbonate PEG. The reversibility of the histidine modification was examined. The PEG-imidazole adduct in the intact protein, PEG-IFN(His-34), is labile but much more stable than in the peptide, PEG-IFN(His-34)dig. Apparently, the tertiary structure of the intact protein protects the His(34)-imidazole ring from depegylation.


Subject(s)
Interferon-alpha/chemistry , Polyethylene Glycols/chemistry , Drug Stability , Histidine/chemistry , Hydrogen-Ion Concentration , Imidazoles/chemistry , Interferon alpha-2 , Interferon-alpha/isolation & purification , Isomerism , Light , Nuclear Magnetic Resonance, Biomolecular , Polyethylene Glycols/isolation & purification , Polymers/chemistry , Recombinant Proteins , Scattering, Radiation
4.
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
5.
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
6.
J Neurochem ; 56(4): 1184-90, 1991 Apr.
Article in English | MEDLINE | ID: mdl-2002336

ABSTRACT

Nigrostriatal cell death in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced Parkinson's disease results from the inhibition of mitochondrial respiration by 1-methyl-4-phenylpyridinium (MPP+). MPP+ blocks electron flow from NADH dehydrogenase to coenzyme Q at or near the same site as do rotenone and piericidin and protects against binding of and loss of activity due to these inhibitors. The 4'-analogs of MPP+ showed increasing affinity for the site with increasing length of alkyl chain, with the lowest Ki, for 4'-heptyl-MPP+, being 6 microM. The 4'-analogs compete with rotenone for the binding site in a concentration-dependent manner. They protect the activity of the enzyme from inhibition by piericidin in parallel to preventing its binding, indicating that the analogs and piericidin bind at the same inhibitory site(s). The optimum protection, however, was afforded by 4'-propyl-MPP+. The lesser protection by the more lipophilic MPP+ analogs with longer alkyl chains may involve a different orientation in the hydrophobic cleft, allowing rotenone and piericidin to still bind even when the pyridinium cation is in a position to interrupt electron flow from NADH to coenzyme Q.


Subject(s)
1-Methyl-4-phenylpyridinium/metabolism , NADH Dehydrogenase/metabolism , Rotenone/metabolism , 1-Methyl-4-phenylpyridinium/analogs & derivatives , 1-Methyl-4-phenylpyridinium/pharmacology , Animals , Binding Sites , Electron Transport , Ions , Multienzyme Complexes/antagonists & inhibitors , NADH, NADPH Oxidoreductases/antagonists & inhibitors , Osmolar Concentration , Pyridines/metabolism
7.
Biochem J ; 273(Pt 2): 481-4, 1991 Jan 15.
Article in English | MEDLINE | ID: mdl-1991043

ABSTRACT

1-Methyl-4-phenylpyridinium (MPP+), the neurotoxic bioactivation product of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), interrupts mitochondrial electron transfer at the NADH dehydrogenase-ubiquinone junction, as do the respiratory chain inhibitors rotenone, piericidin A and barbiturates. Proof that these classical respiratory chain inhibitors and MPP+ react at the same site in the complex NADH dehydrogenase molecule has been difficult to obtain because none of these compounds bind covalently to the target. The 4'-alkyl derivatives of MPP+ inhibit NADH oxidation in submitochondrial particles at much lower concentrations than does MPP+ itself, but still dissociate on washing the membrane preparations, with consequent re-activation of the enzyme. The MPP+ analogues with short alkyl chains prevent the binding of 14C-labelled piericidin A to the membrane and thus must act at the same site, but analogues with alkyl chains longer than heptyl do not prevent binding of [14C]piericidin.


Subject(s)
1-Methyl-4-phenylpyridinium/pharmacology , Mitochondria, Heart/metabolism , Animals , Barbiturates/pharmacology , Carbon Radioisotopes , Cattle , Centrifugation , Electron Transport , Enzyme Activation , Mitochondria, Heart/drug effects , Mitochondria, Heart/enzymology , Multienzyme Complexes/metabolism , NADH Dehydrogenase/metabolism , NADH, NADPH Oxidoreductases/metabolism , Pyridines/pharmacology , Rotenone/pharmacology
8.
Biochem Biophys Res Commun ; 172(3): 1338-41, 1990 Nov 15.
Article in English | MEDLINE | ID: mdl-2244915

ABSTRACT

It is well established that 1-methyl-4-phenylpyridinium (MPP), the neurotoxic bioactivation product of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and most of its analogs are good competitive inhibitors of monoamine oxidase A, with Ki values in the micromolar range, but they inhibit monoamine oxidase B only at much higher concentrations. We report here the finding that alkyl derivatives of MPP+ substituted at the 4' position of the aromatic ring are considerably more effective reversible inhibitors of the A type enzyme, with Ki values in the nanomolar range (0.075-1.6 microM). They inhibit the B type enzyme only at 2 to 3 orders of magnitude higher concentrations (32-374 microM).


Subject(s)
1-Methyl-4-phenylpyridinium/pharmacokinetics , Mitochondria/drug effects , Monoamine Oxidase Inhibitors/pharmacokinetics , Placenta/drug effects , 1-Methyl-4-phenylpyridinium/administration & dosage , 1-Methyl-4-phenylpyridinium/analogs & derivatives , Binding, Competitive , Dose-Response Relationship, Drug , Monoamine Oxidase Inhibitors/administration & dosage
9.
J Med Chem ; 33(8): 2221-30, 1990 Aug.
Article in English | MEDLINE | ID: mdl-2374149

ABSTRACT

The in vivo dopaminergic neurotoxic properties of 45 MPTP and MPP+ analogues and related compounds were examined by an intrastriatal microdialysis assay in conscious rats. MPP(+)-like toxicity, as evidenced by the irreversible effects on DA release and enhancement of lactate formation, was observed with a variety of structural types although no compound was more toxic than MPP+. The following global structure-toxicity relationships could be derived: (1) only permanently charged compounds showed neurotoxic effects; (2) with the exception of amino groups, hydrophilic substituents abolished toxicity; (3) activity was enhanced by lipophilic groups although increased steric bulk around the nitrogen atom tended to decrease activity; (4) nonaromatic, quaternary systems (methiodide of MPTP, guanidinium derivatives) were only weakly toxic; and (5) certain bi- and tricyclic systems, including putative metabolites of potential endogenous MPTP-like compounds, were weakly toxic. The lack of toxic effects following perfusions with DA itself confirmed that MPTP dopaminergic neurotoxicity is not likely to be mediated by the MPP(+)-induced release of DA. With some interesting exceptions, these in vivo data correlate reasonably well with in vitro data on the nerve terminal uptake properties and the inhibitory effects on mitochondrial respiration of these compounds.


Subject(s)
1-Methyl-4-phenylpyridinium/analogs & derivatives , Brain/drug effects , 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 , 1-Methyl-4-phenylpyridinium/toxicity , Animals , Brain/metabolism , Chemical Phenomena , Chemistry , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Dialysis , Dopamine/metabolism , Kinetics , Lactates/metabolism , Lactic Acid , MPTP Poisoning , Male , Molecular Structure , Rats , Rats, Inbred Strains , Structure-Activity Relationship
10.
Biochem Biophys Res Commun ; 169(2): 758-64, 1990 Jun 15.
Article in English | MEDLINE | ID: mdl-2357232

ABSTRACT

1-Methyl-4-phenylpyridinium ion, a major brain metabolite of the neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, is an inhibitor of Complex I of the mitochondrial respiratory chain. We have synthesized several analogs of 1-methyl-4-phenylpyridinium ion containing various alkyl groups in the 4' position of the phenyl ring and have tested them for their abilities to inhibit the oxidation of NADH-linked substrates by intact mouse liver mitochondria. These compounds are considerably more potent inhibitors than MPP+ itself, with potency increasing as the length of the alkyl chain increases. The most potent inhibitor, 1-methyl-4-(4'heptylphenyl)pyridinium ion, was about 200 times as effective as MPP+. These analogs should prove to be useful tools for studying the nature of the process whereby MPP+ and its pyridinium analogs interact with Complex I to inhibit mitochondrial respiration.


Subject(s)
1-Methyl-4-phenylpyridinium/analogs & derivatives , 1-Methyl-4-phenylpyridinium/pharmacology , Mitochondria, Liver/metabolism , Oxygen Consumption/drug effects , Animals , Glutamates/metabolism , Kinetics , Malates/metabolism , Mice , Mitochondria, Liver/drug effects , Molecular Structure , Structure-Activity Relationship
11.
Biochem J ; 268(1): 219-24, 1990 May 15.
Article in English | MEDLINE | ID: mdl-2344359

ABSTRACT

1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and its primary oxidation product, 1-methyl-4-phenyl-2,3-dihydropyridinium (MPDP+), are mechanism-based inhibitors of monoamine oxidases A and B. The pseudo-first-order rate constants for inactivation were determined for various analogues of MPTP and MPDP+ and the concentrations in all redox states were measured throughout the reaction. Disproportionation was observed for all the dihydropyridiniums, but non-enzymic oxidation was insignificant. The dihydropyridiniums were poor substrates for monoamine oxidase A and, consequently, inactivated the enzyme only slowly, despite partition coefficients lower than those for the tetrahydropyridines. For monoamine oxidase B, the dihydropyridiniums were more effective inactivators than the tetrahydropyridines. Substitutions in the aromatic ring had no major effect on the inactivation of monoamine oxidase B, but the 2'-ethyl- and 3'-chloro-substituted compounds were very poor mechanism-based inactivators of monoamine oxidase A. It is clear that both oxidation steps can generate the reactive species responsible for inactivation.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Monoamine Oxidase Inhibitors/pharmacology , Pyridinium Compounds/pharmacology , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/analogs & derivatives , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/metabolism , Female , Humans , Hydrogen-Ion Concentration , Kinetics , Monoamine Oxidase/metabolism , Oxidation-Reduction , Pregnancy , Pyridinium Compounds/metabolism , Spectrophotometry , Substrate Specificity
12.
Proc Natl Acad Sci U S A ; 86(23): 9168-72, 1989 Dec.
Article in English | MEDLINE | ID: mdl-2594758

ABSTRACT

Nineteen structural analogs of 1-methyl-4-phenylpyridinium (MPP+) were studied for their capacity to inhibit the mitochondrial oxidation of NAD+-linked substrates and the aerobic oxidation of NADH in inner membrane preparations from cardiac mitochondria. In the majority of cases, a good correlation was found between the two inhibition effects monitored. A few compounds were effective inhibitors of NADH oxidase but had only marginal effects on mitochondrial respiration. From studies of their accumulation by mitochondria, it appears likely that the latter compounds are not effectively concentrated by intact mitochondria by the electrical gradient and, in part for this reason, cannot reach sufficiently high concentrations at the appropriate binding site of NADH dehydrogenase. In addition, evidence is presented that the penetration of pyridinium analogs to the inhibition site in the NADH dehydrogenase complex may also be rate limiting. The data support the thesis that, for a substituted tetrahydropyridine to be acutely neurotoxic, its pyridinium oxidation product must be actively accumulated in the mitochondria and must inhibit NADH-ubiquinone oxidoreductase in its membrane environment.


Subject(s)
1-Methyl-4-phenylpyridinium/analogs & derivatives , 1-Methyl-4-phenylpyridinium/pharmacology , Mitochondria, Liver/metabolism , Multienzyme Complexes/antagonists & inhibitors , NADH, NADPH Oxidoreductases/antagonists & inhibitors , Oxygen Consumption/drug effects , Animals , Kinetics , Mitochondria, Liver/drug effects , NAD/metabolism , Rats , Rats, Inbred Strains , Structure-Activity Relationship
13.
J Neurochem ; 53(6): 1837-42, 1989 Dec.
Article in English | MEDLINE | ID: mdl-2809594

ABSTRACT

Twenty analogs of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) were tested for their capacity to be oxidized by pure monoamine oxidase-A (MAO-A) prepared from human placenta and pure monoamine oxidase-B (MAO-B) prepared from beef liver. Several of the MPTP analogs were very good substrates for MAO-A, for MAO-B, or for both and had low Km values and high turnover numbers. These values were similar to or even better than those of kynuramine and benzylamine, good substrates for MAO-A and MAO-B, respectively. MPTP had relatively low Km values for oxidation by both MAO-A and MAO-B. In contrast, the turnover number for MPTP oxidation by MAO-B was considerably higher than the value for MAO-A. The corresponding pyridinium species of MPTP and several of the MPTP analogs inhibited MAO-A competitively with Ki values at micromolar concentrations; in contrast the pyridinium species inhibited MAO-B competitively at considerably higher concentrations (i.e., 100 microM or greater Ki values). The data provide information concerning the structural requirements for the oxidation of tetrahydropyridines by MAO-A and MAO-B and the inhibition of these enzymes by pyridiniums.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/analogs & derivatives , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/metabolism , Isoenzymes/metabolism , Monoamine Oxidase Inhibitors/pharmacology , Monoamine Oxidase/metabolism , Animals , Kinetics , Monoamine Oxidase Inhibitors/isolation & purification , Oxidation-Reduction , Structure-Activity Relationship , Substrate Specificity
14.
J Pharmacol Exp Ther ; 249(3): 820-8, 1989 Jun.
Article in English | MEDLINE | ID: mdl-2786564

ABSTRACT

Several analogs of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) were synthesized and compared to MPTP for their ability to be oxidized by monoamine oxidase (MAO) and for their ability to cause nigrostriatal dopaminergic neurotoxicity in mice. Most of the compounds were oxidized by mouse brain MAO, either predominantly by the B-form or by both the A- and B-forms. The MAO-catalyzed oxidation of all of the MAO substrates resulted in the formation of dihydropyridinium intermediates which, in turn, except for the dihydropyridinium of 1-methyl-4-benzyl-1,2,3,6-tetrahydropyridine, formed pyridinium species as the final oxidation product. Nine analogs were found to be neurotoxic; all were oxidized by MAO to pyridinium compounds. However, some non-neurotoxic MPTP analogs were also oxidized by MAO. Neither 1-methyl-4-benzyl-1,2,3,6-tetrahydropyridine nor the compounds which were not substrates for MAO were neurotoxic. Also, the neurotoxicity of all of the compounds tested was blocked by inhibiting either MAO-B, MAO-A or both MAO-B and MAO-A together, indicating that MAO activity was necessary for the neurotoxicity of the compounds to be manifested. The capacity of an MPTP analog to be oxidized by MAO to a pyridinium appears to be a necessary, but not sufficient, parameter in determining the neurotoxic potential of the compound.


Subject(s)
Brain/drug effects , Monoamine Oxidase/metabolism , Pyridines/toxicity , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Animals , Brain/metabolism , Dopamine/metabolism , Drug Interactions , Male , Mice , Monoamine Oxidase Inhibitors/pharmacology , Oxidation-Reduction , Pyridines/metabolism , Structure-Activity Relationship
15.
J Pharmacol Exp Ther ; 249(3): 829-35, 1989 Jun.
Article in English | MEDLINE | ID: mdl-2786565

ABSTRACT

In the accompanying paper, several tetrahydropyridine analogs of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) were screened for their abilities to be oxidized by monoamine oxidase (MAO) to pyridiniums and to produce neurotoxicity in mice. We reported that most of the analogs were oxidized by MAO to pyridiniums and some of the analogs were neurotoxic. We concluded that the capacity of a tetrahydropyridine MPTP analog to be oxidized by MAO to a pyridinium was a necessary, but not sufficient, condition for the compound to be a neurotoxin. In the present paper we attempt to explain further the neurotoxicity or lack of neurotoxicity of these analogs by evaluating the abilities of the pyridinium compounds to serve as substrates for the neostriatal dopamine (DA) transport system and as inhibitors of mitochondrial respiration. We now report that all of the neurotoxic MPTP analogs are oxidized to pyridiniums that are good substrates for the neostriatal DA carrier and good inhibitors of mitochondrial respiration. The results are consistent with an important role for both uptake of the pyridiniums by the DA carrier and inhibition by the pyridiniums of mitochondrial respiration in the neurotoxicity induced by MPTP and its analogs.


Subject(s)
Brain/drug effects , Monoamine Oxidase/metabolism , Pyridines/toxicity , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Animals , Brain/metabolism , Dopamine/metabolism , Dopamine Antagonists , Flunarizine/analogs & derivatives , Flunarizine/pharmacology , Male , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Oxidation-Reduction , Pyridines/metabolism , Pyridinium Compounds/pharmacology , Structure-Activity Relationship
16.
Proc Natl Acad Sci U S A ; 85(16): 6172-6, 1988 Aug.
Article in English | MEDLINE | ID: mdl-3137566

ABSTRACT

1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) is a potent dopaminergic neurotoxin that causes biochemical, pharmacological, and pathological deficits in experimental animals similar to those seen in human parkinsonian patients. All of the deficits can be prevented by treating mice with selective inhibitors of monoamine oxidase B (MAO-B), including deprenyl, prior to MPTP administration. We now report that the dopaminergic neurotoxicity of two potent MPTP analogs, namely the 2'-methyl and 2'-ethyl derivatives (2'-MeMPTP and 2'-EtMPTP), cannot be prevented by deprenyl pretreatment. However, the neurotoxicity of these two analogs can be prevented by pretreatment with a combination of deprenyl and the selective MAO-A inhibitor clorgyline at doses that are sufficient to almost completely inhibit both MAO-B and MAO-A activities. Moreover, the neurotoxicity of 2'-EtMPTP (but not of 2'-MeMPTP and MPTP) can be significantly attenuated by clorgyline alone. There was a parallel between the capacity of the MAO inhibitors to decrease the brain content of the pyridinium species after administration of the tetrahydropyridines and the capacity of the MAO inhibitors to protect against the neurotoxic action of the tetrahydropyridines. The data support the conclusion that both 2'-MeMPTP and 2'-EtMPTP are bioactivated to pyridinium species to a significant extent by MAO-A. Further, it appears that the formation of the pyridinium species plays an important role in the neurotoxic process.


Subject(s)
Monoamine Oxidase/physiology , Pyridines/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Animals , Biotransformation , Brain/metabolism , Clorgyline/pharmacology , Male , Mice , Oxidation-Reduction , Pyridines/toxicity , Selegiline/pharmacology , Structure-Activity Relationship
17.
Toxicology ; 49(2-3): 493-501, 1988 May.
Article in English | MEDLINE | ID: mdl-3259742

ABSTRACT

The nigrostriatal dopaminergic neurotoxicity of MPTP was prevented in mice in a dose-dependent manner by the monoamine oxidase-B (MAO-B) inhibitor deprenyl. This finding, combined with other observations, points out the important role of MAO-B in the bioactivation of MPTP. In the present study, some comparisons between MPTP and several of its structural analogs will be presented.


Subject(s)
Pyridines/toxicity , Receptors, Dopamine/drug effects , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , 1-Methyl-4-phenylpyridinium , Animals , Dose-Response Relationship, Drug , Male , Mice , Monoamine Oxidase Inhibitors/pharmacology , Pyridinium Compounds/toxicity , Structure-Activity Relationship
18.
Eur J Pharmacol ; 146(2-3): 313-8, 1988 Feb 09.
Article in English | MEDLINE | ID: mdl-3131149

ABSTRACT

1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) administration leads to the selective destruction of the dopaminergic neurons of the nigrostriatal pathway in experimental animals including monkeys and mice. The neurotoxicity of MPTP is dependent upon its monoamine oxidase-B (MAO-B)-catalyzed conversion to the 1-methyl-4-phenylpyridinium species (MPP+). A methylated analog of MPTP. A methylated analog of MPTP, namely 1-methyl-4-(2'-methylphenyl)-1,2,3,6-tetrahydropyridine (2'Me-MPTP), is a more potent dopaminergic neurotoxin than MPTP in mice. Although the selective inhibition of MAO-B is sufficient to protect mice against MPTP-induced neurotoxicity, it is reported here that complete inhibition of MAO-B failed to prevent 2'Me-MPTP-induced dopaminergic neurotoxicity. However, the neurotoxicity of 2'Me-MPTP was completely prevented and 2'Me-MPP+ formation was markedly attenuated in mice in which both MAO-A and MAO-B were almost totally inhibited. This information about the role of MAO-A in the bioactivation of 2'Me-MPTP may be of relevance to those who speculate that the MAO-B catalyzed bioactivation of MPTP or a similar compound may be the cause of idiopathic Parkinson's disease.


Subject(s)
Corpus Striatum/drug effects , Dopamine/metabolism , Monoamine Oxidase/physiology , Neurotoxins/toxicity , Pyridines/toxicity , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Animals , Biotransformation , Clorgyline/pharmacology , Corpus Striatum/metabolism , Haplorhini , Mice , Monoamine Oxidase Inhibitors/pharmacology , Neural Pathways/drug effects , Neurotoxins/metabolism , Pyridines/metabolism , Selegiline/pharmacology
19.
J Pharmacol Exp Ther ; 242(3): 850-7, 1987 Sep.
Article in English | MEDLINE | ID: mdl-2888874

ABSTRACT

1-Methyl-4-(2'-methylphenyl)-1,2,3,6-tetrahydropyridine (2'CH3-MPTP) was shown previously to be a more potent neurotoxicant than 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) in mice. The present investigation was conducted to determine possible reasons for the greater potency of 2'CH3-MPTP and to determine if its neurotoxic action might be similar to that of MPTP. 2'CH3-MPTP was a much better substrate for monoamine oxidase than was MPTP (Km values of 66 and 114 microM and Vmax values of 3433 and 1389 nmol/g of tissue per hr for 2'CH3-MPTP and MPTP, respectively) and it is likely that this is an important feature which contributes to its greater potency. In addition, its pyridinium metabolite, 1-methyl-4-(2'-methylphenyl)pyridinium was found to be an excellent substrate for the dopamine carrier with Km and Vmax values (513 nM and 4.1 nmol/g of tissue per min, respectively) similar to those of 1-methyl-4-phenylpyridinium (872 nM and 5.2 nmol/g of tissue per min, respectively). In vivo, 2'CH3-MPTP-induced neurotoxicity, like MPTP-induced neurotoxicity, was attenuated by the pretreatment of mice with a dopamine uptake inhibitor (mazindol or GBR 13069). However, selective doses of the monoamine oxidase (MAO)-B inhibitors, deprenyl or MDL 72145, failed to prevent in vivo neurotoxicity induced by 2'CH3-MPTP although these doses effectively blocked MPTP-induced neurotoxicity. Protection against 2'CH3-MPTP-induced neurotoxicity was observed only at a nonselective dose of MDL 72145 which blocked both MAO-B and MAO-A activities.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Brain/drug effects , Pyridines/toxicity , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Animals , Brain/metabolism , Dopamine/analysis , Dose-Response Relationship, Drug , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Monoamine Oxidase Inhibitors/pharmacology , Neurotransmitter Uptake Inhibitors/pharmacology , Oxidation-Reduction , Pyridines/metabolism , Pyridines/pharmacokinetics , Tyrosine 3-Monooxygenase/analysis
20.
Neurosci Lett ; 79(1-2): 151-6, 1987 Aug 18.
Article in English | MEDLINE | ID: mdl-3499585

ABSTRACT

1-Methyl-4-cyclohexyl-1,2,3,6-tetrahydropyridine (MCTP), an analog of MPTP, was found to be an MPTP-like neurotoxin. MCTP administration caused extensive losses of neostriatal dopamine and its major metabolites in male Swiss-Webster mice. Under similar experimental conditions, MCTP was approximately as potent as MPTP. Like MPTP, MCTP was a good substrate for monoamine oxidase-B (MAO-B) and its neurotoxicity was prevented in mice by AGN-1135, a selective inhibitor of MAO-B. The neurotoxicity of MCTP and of MPTP was also prevented by the dopamine uptake inhibitor mazindol. 1-Methyl-4-cyclohexylpyridinium ion (MCP+), the 4-electron oxidation product of MCTP, caused release of previously accumulated [3H]dopamine from mouse neostriatal synaptosomes. This release was blocked by mazindol, which indicates that MCP+, like 1-methyl-4-phenylpyridinium ion (MPP+), the 4-electron oxidation product of MPTP, is a substrate for the dopamine transport system. Like MPP+, MCP+ was found to inhibit the mitochondrial oxidation of NADH-linked substrates. It appears that conjugation between the tetrahydropyridine ring and a 4-substituent is not a requirement for an MPTP analog to possess neurotoxicity.


Subject(s)
Caudate Nucleus/drug effects , Putamen/drug effects , Pyridines/toxicity , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Animals , Caudate Nucleus/metabolism , Dopamine/metabolism , Indans/pharmacology , Male , Mazindol/pharmacology , Mice , Mice, Inbred BALB C , Putamen/metabolism , Synaptosomes/drug effects , Synaptosomes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...