Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
2.
Life Sci ; 110(1): 1-7, 2014 Aug 06.
Article in English | MEDLINE | ID: mdl-24880075

ABSTRACT

AIMS: The molecular mechanisms for the loss of 3,4-dihydroxyphenylalanine (l-dopa) efficacy during the treatment of Parkinson's disease (PD) are unknown. Modifications related to catecholamine metabolism such as changes in l-dopa and dopamine (DA) metabolism, the modulation of catecholamine enzymes and the production of interfering metabolites are the primary concerns of this study. MAIN METHODS: Normal (saline) and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) pre-treated mice were primed with 100mg/kg of l-dopa twice a day for 14 days, and a matching group remained l-dopa naïve. l-dopa naive and primed mice received a challenge dose of 100mg/kg of l-dopa and were sacrificed 30 min later. Striatal catecholamine levels and the expression and activity of catechol-O-methyltransferase (COMT) were determined. KEY FINDINGS: Normal and MPTP pre-treated animals metabolize l-dopa and DA similarly during l-dopa therapy. Administration of a challenge dose of l-dopa increased l-dopa and DA metabolism in l-dopa naïve animals, and this effect was enhanced in l-dopa primed mice. The levels of 3-OMD in MPTP pre-treated animals were almost identical to those in normal mice, which we found are likely due to increased COMT activity in MPTP pre-treated mice. SIGNIFICANCE: The results of this comparative study provide evidence that sub-chronic administration of l-dopa decreases the ability of the striatum to accumulate l-dopa and DA, due to increased metabolism via methylation and oxidation. This data supports evidence for the metabolic adaptation of the catecholamine pathway during long-term treatment with l-dopa, which may explain the causes for the loss of l-dopa efficacy.


Subject(s)
Antiparkinson Agents/pharmacology , Catechol O-Methyltransferase/metabolism , Dopamine/metabolism , Levodopa/pharmacology , Parkinsonian Disorders/drug therapy , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/administration & dosage , Animals , Antiparkinson Agents/administration & dosage , Corpus Striatum/metabolism , Disease Models, Animal , Drug Administration Schedule , Levodopa/administration & dosage , Male , Methylation , Mice , Mice, Inbred C57BL , Oxidation-Reduction , Parkinsonian Disorders/physiopathology , Time Factors
3.
Life Sci ; 91(19-20): 921-7, 2012 Nov 02.
Article in English | MEDLINE | ID: mdl-23000099

ABSTRACT

AIMS: In this study, the effects of prenatal exposure to methanol (MeOH) on the nigrostriatal dopamine (NSDA) system were examined to determine if the interaction could sensitize this system, and serve as an underpinning for Parkinson's disease (PD) like changes that occur later in life. Methanol was studied because its toxicity resembles the symptoms of PD and the symptoms are relieved by L-dopa meaning that MeOH targets the NSDA system. Since fermentation and wood combustion are major sources for MeOH, the incidence of human encounters with MeOH is high. As a superior solvent and the precursor for formaldehyde, MeOH has a powerful and sometimes, irreversible impact on chemical processes, such as cross-linking proteins and nucleic acids. It may cause subthreshold changes that sensitizes the NSDA system to PD, that occur during aging. MAIN METHODS: To study the prenatal effects of MeOH, pregnant C57BL/6J mice were administered 40 mg/kg MeOH by oral gavage during gestation days 8-12, twice daily. Twelve weeks after birth, behavior impairments were recorded. The striatum was dissected for the determination of tyrosine hydroxylase (TH), L-aromatic amino acid decarboxylase (LAAD), α-synuclein and levels of dopamine (DA) and its metabolites. KEY FINDINGS: MeOH reduced striatal TH and LAAD protein by 47% and 57% respectively and DA by 32%. SIGNIFICANCE: The results mean that in utero exposure to toxins similar to MeOH could sensitize the striatal system to changes that cause PD. This study may help identify strategies to block this type of in utero toxicity.


Subject(s)
Dopamine/metabolism , Methanol/toxicity , Parkinson Disease/etiology , Prenatal Exposure Delayed Effects , Animals , Aromatic-L-Amino-Acid Decarboxylases/metabolism , Behavior, Animal/drug effects , Corpus Striatum/metabolism , Disease Models, Animal , Female , Male , Methanol/administration & dosage , Mice , Mice, Inbred C57BL , Parkinson Disease/physiopathology , Pregnancy , Tyrosine 3-Monooxygenase/metabolism , alpha-Synuclein/metabolism
4.
Pharmacol Biochem Behav ; 83(3): 349-59, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16580056

ABSTRACT

Although the etiology of Parkinson's disease (PD) is not fully understood, there are numerous studies that have linked the increased risk for developing PD to pesticides exposure including paraquat (PQ). Moreover, the exposure to a combination of compounds or chemical mixtures has been suggested to further increase this risk. In the current study, the effects of PQ on the nigrostriatal dopaminergic system in male C57BL6 mice exposed to 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) were examined to assess the impact of toxic substance mixtures exposure on neurochemical and behavioral changes. In this study, a low non-toxic dose of MPTP (10mg/kg) was injected once a day for 5 days and was followed by PQ (7 mg/kg) once a day for 6 days (subacute protocol) or once a week for 10 weeks (chronic protocol). The results from the subacute protocol showed that PQ reduced the turnover of dopamine (DA) as indicated by a 21% and a 22.3% decrease in dihydroxyphenyl acetic acid (DOPAC), homovanillic acid and increased S-adenosyl methionine/S-adenosyl homocysteine index (SAM/SAH) by 100%. However, the administration of PQ to MPTP primed mice resulted in the decrease of DOPAC, HVA, DA, by 35.8%, 35.2% and 22.1%, respectively. In addition, PQ decreased the total number of movements (TM) by 28% but MPTP plus PQ decreased TM by 41%. The SAM/SAH index showed that MPTP increased methylation by 33.3%, but MPTP plus PQ increased methylation by 81%. In the chronic protocol, the data showed that MPTP administration did not affect DA, DOPAC, and HVA levels. The administration of PQ led to significant decrease in DOPAC, HVA, and TD by 31.6%, 19.9%, and 21.2% respectively with no effect on DA levels. The MPTP plus PQ group showed reduced DA, DOPAC, HVA, and total distance traveled by 58.4%, 82.8%, 55.8%, and 83.9%, respectively. Meanwhile, PQ administration caused a reduction in tyrosine hydroxylase immunoreactivity in the substantia nigra, and this effect was more pronounced in MPTP pretreated mice. It was concluded from this study that prior treatment with MPTP potentiated the effects of PQ in reducing DA, DOPAC, HVA, TH immunoreactivity, locomotor activity, and increasing the methylation index. The enhanced effects of PQ following MPTP administration further support the role of toxic substance mixtures in causing Parkinson's disease.


Subject(s)
Behavior, Animal/drug effects , MPTP Poisoning/etiology , MPTP Poisoning/psychology , Neurotoxins/toxicity , Paraquat/toxicity , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/administration & dosage , 3,4-Dihydroxyphenylacetic Acid/metabolism , Animals , Dopamine/chemistry , Dopamine/metabolism , Drug Synergism , Herbicides/administration & dosage , Herbicides/toxicity , Homovanillic Acid/metabolism , Humans , MPTP Poisoning/metabolism , Male , Methylation , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Neurotoxins/administration & dosage , Paraquat/administration & dosage
5.
Neurotoxicology ; 26(6): 945-57, 2005 Dec.
Article in English | MEDLINE | ID: mdl-15950286

ABSTRACT

Excessive methylation has been proposed to be involved in the pathogenesis of Parkinson's disease (PD), via mechanisms that involve phospholipid methylation. Meanwhile, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) was found to stimulate phospholipid methylation via the oxidized metabolite, 1-methyl-4-phenyl-pyridinium (MPP+), in the rat brain and liver tissues. In the present study, we investigated the effect of MPP+ on phosphatidylethanolamine N-methyltransferases (PENMT) and the potential role of this pathway in MPP(+)-induced neurotoxicity using PC12 cells. The results obtained indicate that MPP+ stimulated phosphatidylethanolamine (PTE) methylation to phosphatidylcholine (PTC) and correspondingly increased the formation of lysophosphatidylcholine (lyso-PTC). Moreover, the addition of S-adenosylmethionine (SAM) to the cell culture medium increases MPP(+)-induced cytotoxicity. The incubation of 1mM MPP+ and various concentrations of SAM (0-4 mM) decreased the viability of PC12 cells from 80% with MPP+ alone to 38% viability with 4 mM SAM for 4 days incubation. The data also revealed that the addition of S-adenosylhomocysteine (SAH), a methylation inhibitor, offered significant protection against MPP(+)-induced cytotoxicity, indicating that methylation plays a role in MPP(+)-induced cytotoxicity. Interestingly, lyso-PTC showed similar actions to MPP+ in causing many cytotoxic changes with at least 10 times higher potency. Lyso-PTC induced dopamine release and inhibited dopamine uptake in PC12 cells. Lyso-PTC also caused the inhibition of mitochondrial potential and increased the formation of reactive oxygen species in PC12 cells. These results indicate that phospholipid methylation pathway might be involved in MPP+ neurotoxicity and lyso-PTC might play a role in MPP(+)-induced neurotoxicity.


Subject(s)
Lysophosphatidylcholines/biosynthesis , MPTP Poisoning/metabolism , Phosphatidylethanolamine N-Methyltransferase/drug effects , Phospholipids/metabolism , 1-Methyl-4-phenylpyridinium , Animals , Dopamine/metabolism , Lysophosphatidylcholines/metabolism , Methylation/drug effects , PC12 Cells , Phosphatidylcholines/biosynthesis , Phosphatidylethanolamine N-Methyltransferase/metabolism , Phosphatidylethanolamines/metabolism , Rats , S-Adenosylhomocysteine/pharmacology , S-Adenosylmethionine/pharmacology
6.
Neurotoxicology ; 26(3): 361-71, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15935208

ABSTRACT

Long-term treatment of levodopa for Parkinson's disease (PD) patients is known to elevate homocysteine level in their plasma. The present study was designed to examine the possible neurotoxic effects of the increased homocysteine level on the dopaminergic system. Homocysteine was administered into Sprague-Dawley male rats intracerebroventricularly or C57BL/6 mice intraperitoneally. Following homocysteine injection the locomotor activities, the levels of dopamine (DA) and its metabolites, 3,4-dihydroxyphenylacetic acid (DOPAC) and homovanillic acid (HVA), and immunohistochemistry of dopaminergic neurons were examined. The results obtained indicate that homocysteine administration (1 or 2 micromol, i.c.v.) into the rat brains for 5 days significantly decreased the locomotor activities and dopamine as well as its metabolites, DOPAC and HVA, in the rat striatal regions. Two different doses of homocysteine (50 and 100mg/100g, i.p. daily) were administered into mice for 36 days to evaluate the effect of systemic treatment of homocysteine on the dopaminergic neurons of the brain. The intraperitoneal injections of two doses of homocysteine significantly increased homocysteine levels in the striatal regions of mouse brains by 21.5 and 39.2%, while reducing dopamine turnover rates in the striatal regions by decreasing (DOPAC+HVA)/DA, 23.7 and 51.6%, respectively. Accordingly, homocysteine decreased locomotor activities significantly by decreasing movement time by 29 and 38%, total distance by 32 and 42%, and numbers of movement by 28 and 41%, respectively. Moreover, homocysteine decreased tyrosine hydroxylase immunoreactivity in substantia nigra of mouse brain. The data obtained indicate that the potential of homocysteine to be toxic to the dopaminergic system. Consequently, long-term levodopa therapy for PD may accelerate the progression of PD, at least in part by elevated homocysteine.


Subject(s)
Behavior, Animal/drug effects , Dopamine/physiology , Homocysteine/toxicity , Neurotoxicity Syndromes/psychology , Tyrosine/analogs & derivatives , Adenosine/pharmacology , Animals , Brain Chemistry/drug effects , Catheterization , Chromatography, High Pressure Liquid , Dopamine/metabolism , Homocysteine/administration & dosage , Immunohistochemistry , Injections, Intraventricular , Male , Methylation , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Neostriatum/drug effects , Neostriatum/metabolism , Neurotoxicity Syndromes/physiopathology , Rats , Rats, Sprague-Dawley , Substantia Nigra/enzymology , Substantia Nigra/metabolism , Tyrosine/pharmacology , Tyrosine 3-Monooxygenase/metabolism
7.
J Biol Chem ; 280(1): 677-84, 2005 Jan 07.
Article in English | MEDLINE | ID: mdl-15504733

ABSTRACT

(S)-adenosylmethionine (SAM) is a critical element of melatonin synthesis as the methyl donor in the last step of the pathway, the O-methylation of N-acetyl 5-hydroxytryptamine by hydroxyindole-O-methyltransferase. The activity of the enzyme that synthesizes SAM, methionine adenosyltransferase (MAT), increases 2.5-fold at night in the pineal gland. In this study, we found that pineal MAT2A mRNA and the protein it encodes, MAT II, also increase at night, suggesting that the increase in MAT activity is caused by an increase in MAT II gene products. The night levels of MAT2A mRNA in the pineal gland were severalfold higher than in other neural and non-neural tissues examined, consistent with the requirement for SAM in melatonin synthesis. Related studies indicate that the nocturnal increase in MAT2A mRNA is caused by activation of a well described neural pathway that mediates photoneural-circadian regulation of the pineal gland. MAT2A mRNA and MAT II protein were increased in organ culture by treatment with norepinephrine (NE), the sympathetic neurotransmitter that stimulates the pineal gland at night. NE is known to markedly elevate pineal cAMP, and here it was found that cAMP agonists elevate MAT2A mRNA levels by increasing MAT2A mRNA synthesis and that drugs that block cAMP activation of cAMP dependent protein kinase block effects of NE. Therefore, the NE-cAMP dependent increase in pineal MAT activity seems to reflect an increase in MAT II protein, which occurs in response to cAMP-->protein kinase-dependent increased MAT2A expression. The existence of this MAT regulatory system underscores the importance that MAT plays in melatonin biogenesis. These studies also point to the possibility that SAM production in other tissues might be regulated through cAMP.


Subject(s)
Cyclic AMP/metabolism , Gene Expression Regulation, Enzymologic , Methionine Adenosyltransferase/metabolism , Pineal Gland/enzymology , Animals , Circadian Rhythm , Male , Methionine Adenosyltransferase/genetics , Organ Specificity , Pineal Gland/physiology , Rats , Rats, Sprague-Dawley
8.
Neurotoxicology ; 26(1): 27-38, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15527871

ABSTRACT

Lysophosphatidylcholine (lyso-PTC), a secondary product of S-adenosylmethionine (SAM)-dependent phosphatidylethanolamine (PTE) methylation, is a potent cytotoxin and might be involved in the pathogenesis of Parkinson's disease (PD). Our previous studies showed that the injection of SAM into the brain caused PD-like changes in rodents. Moreover, 1-methyl-4-phenylpyridinium (MPP+), a Parkinsonism-inducing agent, increased lyso-PTC formation via the stimulation of PTE methylation pathway. These results indicate a possible role of lyso-PTC in the PD-like changes seen following the injection of SAM or MPP+. In the present study, lyso-PTC was injected into the lateral ventricle of rats and locomotor activities and the biogenic amine levels were measured to evaluate the effects of lyso-PTC on the dopaminergic system. Quinacrine, a phospholipase A2 (PLA2) inhibitor, was employed to determine its protective effect on SAM-induced PD-like changes by the inhibition of lyso-PTC formation. The results showed that 1 h after the injection, 0.4 and 0.8 micromol of lyso-PTC increased striatal dopamine (DA) by 20 and 24%, decreased 3,4-dihydroxyphenylacetic acid (DOPAC) by 37 and 45% and decreased homovanilic acid (HVA) by 24 and 13%, respectively. Consequently, dopamine turnover rate, (DOPAC + HVA)/DA, was significantly reduced by 44 and 48% in the rat striatum. Meanwhile, the administration of 0.4 or 0.8 micromol of lyso-PTC decreased movement time by 52 and 63%, total distance by 44 and 48% and the number of movements by 43 and 64%, respectively. Quinacrine attenuated SAM-induced hypokinesia without affecting SAM metabolism prior to its action on rat brain. The results obtained indicate that the hypokinesia observed following the administration of lyso-PTC might be related to the decline in DA turnover in the striatum in response to lyso-PTC exposure. The present study suggests that inhibitory effects of lyso-PTC on dopaminergic neurotransmission is one of the contributing factors in SAM and MPP+-induced PD-like changes.


Subject(s)
Dopamine/metabolism , Lysophosphatidylcholines/pharmacology , Motor Activity/drug effects , Animals , Brain Chemistry/drug effects , Chromatography, High Pressure Liquid , Chromatography, Thin Layer , Hypokinesia/chemically induced , Hypokinesia/physiopathology , Kinetics , Liver/metabolism , Lysophosphatidylcholines/biosynthesis , Male , Methyltransferases/metabolism , Phosphatidylethanolamine N-Methyltransferase , Pyridinium Compounds/pharmacology , Quinacrine/pharmacology , Rats , Rats, Sprague-Dawley , S-Adenosylmethionine/pharmacology , Serotonin/metabolism
9.
Neurochem Res ; 29(7): 1333-42, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15202762

ABSTRACT

Lysophosphatidylcholine (lyso-PTC) is formed by phospholipase A2 (PLA2) from phosphatidylcholine (PTC), that is produced through phosphatidylethanolamine (PTE) methylation. 1-Methyl-4-phenyl-pyridinium (MPP+), a Parkinson's disease (PD) inducing agent, and S-adenosylmethionine (SAM), a biological methyl donor, increase lyso-PTC formation and both induce PD-like changes in animal models. In the current study, we investigated the effect of lyso-PTC on the dopaminergic system to determine the modulating role of lyso-PTC in dopaminergic neurotransmission. The results of these experiments show that lyso-PTC has a remarkable inhibitory effect on dopamine D1 and D2 receptor binding activities in the striatal membrane prepared from Sprague-Dawley rats. Lyso-PTC decreased the Bmax values of both D1 and D2 receptor binding activities. The Kd values for D1 and D2 receptors were not changed, but lyso-PTC also inhibited dopamine transporter and decreased striatal dopamine turnover rate. MPP+ showed similar, but less potent effects. The current studies suggest that lyso-PTC significantly impair the dopaminergic system and might play a role in MPP+ and SAM induced PD-like changes through its inhibitory effects on dopaminergic neurotransmission.


Subject(s)
Dopamine Antagonists/pharmacology , Dopamine/physiology , Lysophosphatidylcholines/pharmacology , Animals , Cell Membrane/metabolism , Corpus Striatum/metabolism , Kinetics , Male , Phospholipids/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Dopamine D1/drug effects , Receptors, Dopamine D1/physiology , Receptors, Dopamine D2/drug effects , Receptors, Dopamine D2/physiology
10.
Neurosci Res ; 48(3): 335-44, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15154679

ABSTRACT

Excess methylation has been suggested to play a role in the pathogenesis of Parkinson's disease (PD), since the administration of S-adenosylmethionine (SAM), a biological methyl donor, induces PD-like changes in rodents. It was proposed that SAM-induced PD-like changes might be associated with its ability to react with the dopaminergic system. In the present study the effects of SAM on dopamine receptors and transporters were investigated using rats and cloned dopamine receptor proteins. Autoradiographic examination of SAM indicated its tendency to be localized and accumulated in rat striatal region after the intracerebroventricular injection into rat brain. Moreover, results showed that SAM significantly decreased dopamine D1 and D2 receptor binding activities by decreasing the Bmax and increasing the Kd values. At concentrations of 0.1, 0.25 and 0.5 mM, SAM was able to reduce the Bmax from the control value of 848.1 for dopamine D1-specific ligand [3H] SCH 23390 to 760.1, 702.6 and 443.0 fmol/mg protein, respectively. At the same concentrations, SAM was able to increase the Kd values from 0.91 for the control to 1.06, 3.84 and 7.01 nM of [3H] SCH 23390, respectively. The effects of SAM on dopamine D2 binding were similar to those of dopamine D1 binding. SAM also decreased dopamine transporter activity. The interaction of SAM with dopamine receptor proteins produced methanol from methyl-ester formation and hydrolysis. We propose that the SAM effect might be related to its ability to react with dopamine receptor proteins through methyl-ester formation and methanol production following the hydrolysis of the carboxyl-methylated receptor proteins.


Subject(s)
Brain/metabolism , Membrane Glycoproteins , Membrane Transport Proteins/metabolism , Nerve Tissue Proteins/metabolism , Receptors, Dopamine/metabolism , S-Adenosylmethionine/pharmacokinetics , Analysis of Variance , Animals , Benzamides/pharmacokinetics , Benzazepines/pharmacokinetics , Binding, Competitive/physiology , Brain/anatomy & histology , Brain/drug effects , Brain Chemistry , Dizocilpine Maleate/pharmacokinetics , Dopamine Antagonists/pharmacokinetics , Dopamine Plasma Membrane Transport Proteins , Dopamine Uptake Inhibitors/pharmacokinetics , Dose-Response Relationship, Drug , Excitatory Amino Acid Antagonists/pharmacokinetics , Injections, Intraventricular/methods , Male , Mazindol/pharmacokinetics , Methylation , Protein Binding/drug effects , Protein Binding/physiology , Radioligand Assay/methods , Rats , Rats, Sprague-Dawley , Tissue Distribution , Tritium/pharmacokinetics
11.
Pharmacol Biochem Behav ; 76(3-4): 433-42, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14643842

ABSTRACT

We previously showed that S-adenosylmethionine (SAM) induces movement impairments similar to those observed in Parkinson's disease (PD) apparently by prenylated protein methylation; 5 kDa molecules being methylated and the symptoms being inhibited by prenylcysteine (PC) analogs. In the present study, we explore the biochemical mechanism of action of the PC analogs. N-acetylgeranylcysteine (AGC), N-acetylfarnesylcysteine (AFC), N-acetylgeranylgeranylcysteine (AGGC), farnesylthioacetic acid (FTA), farnesyl-2-ethanesulfonic acid (FTE) and farnesylsuccinic acid (FMS), but not farnesylthiotriazole (FTT) and farnesylthiolactic acid (FTL), inhibited the SAM-induced motor impairments. Incubation of the respective analogs with rat brain membranes containing prenylated protein methyltransferase (PPMTase) resulted in the methylation of AGC, AFC and AGGC. FTA, FTE, FMS and FTT, but not FTL, inhibited the enzyme activity. A single injection of the active analogs remained effective for at least 3 days against repeated injections of 1 micromol SAM. Amphetamine-induced hyperactivity in rats was inhibited by SAM but potentiated by FTE. During 60 min, the movement time for amphetamine-treated rats was 1477 s compared with 633 and 1664 s for amphetamine+SAM- and amphetamine+FTE-treated rats, respectively. The total distance for amphetamine+FTE-treated rats was 82% higher than for amphetamine. The horizontal activity was 30,728 (amphetamine), 15,430 (FTE), 18,526 (amphetamine+SAM), 41,736 (amphetamine+FTE) and 7004 (SAM) as compared to the PBS control (4726). The intricate relationship between the actions of SAM, which speeds up prenylated protein methylation and impairs movement, amphetamine, which increases synaptic dopamine levels and movement, and the PC analogs, which prevent the SAM-induced movement impairments, suggests a SAM-induced defect on dopamine signaling as the likely cause of the symptoms. The data reveal that interaction of PC analogs with PPMTase may not be an indicator of anti-PD-like activity.


Subject(s)
Cystine/analogs & derivatives , Dyskinesia, Drug-Induced/prevention & control , S-Adenosylmethionine/pharmacology , Amphetamine/antagonists & inhibitors , Amphetamine/pharmacology , Animals , Body Temperature/drug effects , Central Nervous System Stimulants/antagonists & inhibitors , Central Nervous System Stimulants/pharmacology , Cystine/pharmacology , Drug Synergism , Enzyme Inhibitors/pharmacology , Hyperkinesis/chemically induced , Hyperkinesis/prevention & control , In Vitro Techniques , Male , Methylation , Protein Methyltransferases/antagonists & inhibitors , Protein Methyltransferases/metabolism , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , Triazoles/pharmacology
12.
J Neurosci Res ; 69(4): 519-29, 2002 Aug 15.
Article in English | MEDLINE | ID: mdl-12210845

ABSTRACT

Our previous studies showed that S-adenosyl-methionine (SAM) induced Parkinson's disease-like changes in rat. It caused death to dopamine neurons in the substantia nigra, which appeared shrunken and fragmented, indicative of apoptosis-like changes (Charlton and Crowell [1995] Mol. Chem. Neuropathol. 26:269-284; Charlton [1997] Life Sci. 61:495-502). In this study, we investigated whether SAM causes apoptosis in both undifferentiated PC12 (PC12) cells and nerve growth factor (NGF)-differentiated PC12 (D-PC12) cells. S-adenosyl-homocysteine (SAH), the nonmethyl analog of SAM, was also tested. SAM and SAH (1.0 nM to 10.0 microM) caused lactate dehydrogenase (LDH) release from the PC12 cells and D-PC12 cells; cells with morphological changes and fluorescent DNA fragmentation staining were detected among both PC12 cell and D-PC12 cell. Compared with the PC12 cell, the D-PC12 cell, a postmitotic cell, was more sensitive to the toxic effects of SAM or SAH and presented much greater LDH release, suggesting a lethal effect; surprisingly, the amounts of apoptotic cells did not differ significantly between the two kinds of cells. In medium deprived of exogenous methionine, a decline in LDH release was observed in PC12 and D-PC12 cells. Also, lower levels of intracellular SAM and SAH were observed in the methionine-deleted media, which were reversed by the addition of either SAM or SAH. An antivitamin B(12) monoclonal antibody was added to methionine-depleted medium, resulting in deficiency of both endogenous and exogenous methionine, which caused further decreases in LDH release and reduction in the levels of intracellular SAM and SAH. The preliminary data showed different sensitivities to SAM or SAH between PC12 cell and D-PC12 cells, which suggests that PC12 cell may be more stable as a metabolic model. Apoptosis of PC12 cells was also assessed by PARP cleavage detection, Western blot analysis of Bax and Bcl-2 proteins, and DNA laddering on agarose gel electrophoresis. The proapoptoic protein Bax was dominantly expressed, whereas Bcl-2 was slightly down-regulated by SAM. SAH weakly induced the expression of Bax and slightly decreased Bcl-2 levels. The effects of SAM and its analog, SAH, were demonstrated conclusively to induce apoptosis in PC12 cells.


Subject(s)
Apoptosis/drug effects , Brain/metabolism , Cell Differentiation/drug effects , Methionine/deficiency , Neurons/drug effects , Neurons/metabolism , Parkinsonian Disorders/chemically induced , S-Adenosylmethionine/pharmacology , Animals , Apoptosis/physiology , Brain/physiopathology , Cell Differentiation/physiology , Culture Media, Conditioned/pharmacology , DNA Fragmentation/drug effects , DNA Fragmentation/physiology , Intracellular Fluid/drug effects , Intracellular Fluid/metabolism , L-Lactate Dehydrogenase/metabolism , Neurons/pathology , PC12 Cells , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/physiopathology , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases , Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , S-Adenosylhomocysteine/metabolism , S-Adenosylhomocysteine/pharmacology , S-Adenosylmethionine/metabolism , bcl-2-Associated X Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...