Korean J Physiol Pharmacol.  2013 Feb;17(1):89-97. 10.4196/kjpp.2013.17.1.89.

Differential Effects of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine on Motor Behavior and Dopamine Levels at Brain Regions in Three Different Mouse Strains

Affiliations
  • 1Department of Pharmacology, College of Medicine, Dankook University, Cheonan 330-714, Korea. hrkim@dankook.ac.kr

Abstract

Developing an animal model for a specific disease is very important in the understanding of the underlying mechanism of the disease and allows testing of newly developed new drugs before human application. However, which of the plethora of experimental animal species to use in model development can be perplexing. Administration of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) is a very well known method to induce the symptoms of Parkinson's disease in mice. But, there is very limited information about the different sensitivities to MPTP among mouse strains. Here, we tested three different mouse strains (C57BL/6, Balb-C, and ICR) as a Parkinsonian model by repeated MPTP injections. In addition to behavioral analysis, endogenous levels of dopamine and tetrahydrobiopterin in mice brain regions, such as striatum, substantia nigra, and hippocampus were directly quantified by liquid chromatography-tandem mass spectrometry. Repeated administrations of MPTP significantly affected the moving distances and rearing frequencies in all three mouse strains. The endogenous dopamine concentrations and expression levels of tyrosine hydroxylase were significantly decreased after the repeated injections, but tetrahydrobiopterin did not change in analyzed brain regions. However, susceptibilities of the mice to MPTP were differed based on the degree of behavioral change, dopamine concentration in brain regions, and expression levels of tyrosine hydroxylase, with C57BL/6 and Balb-C mice being more sensitive to the dopaminergic neuronal toxicity of MPTP than ICR mice.

Keyword

Dopamine; Mass spectrometry; Mouse strains; MPTP; Parkinson's disease

MeSH Terms

1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine
Animals
Biopterin
Brain
Dopamine
Dopaminergic Neurons
Hippocampus
Humans
Mass Spectrometry
Mice
Mice, Inbred ICR
Models, Animal
Parkinson Disease
Substantia Nigra
Tyrosine 3-Monooxygenase
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine
Biopterin
Dopamine
Tyrosine 3-Monooxygenase

Figure

  • Fig. 1 Effects of MPTP (30 mg/kg, s.c) on Rota-rod performance. Animals were trained to stay on an operating Rota-rod for 600 seconds. On the test day, animals were evaluated on the Rota-rod 6 and 24 hours after MPTP administration. (A) MPTP (C57BL/6, 30 mg/kg, s.c.), (B) MPTP (ICR, 30 mg/kg, s.c.), (C) MPTP (C57BL/6, ICR, Balb C, 20 mg/kg, s.c.), (D) body weight after MPTP administration. Data are expressed as mean latency to fall off the Rota-rod (vertical bars represent standard error of the mean). *p<0.05, **p<0.01 vs 0 hour (naïve) group, #p<0.05 compared between 6 hours and 24 hours groups by Student's t-test, n=6~10.

  • Fig. 2 Summary of experimental design. Each groups received MPTP treatment for 3 days followed by one MPTP-free day. At the end of behavioral test of day 4, animals were euthanized for brain sample collection.

  • Fig. 3 Effect of MPTP (20 mg/kg, s.c) on moving distance for 1 hour following MPTP administration. (A) C57BL/6 mice, (B) Balb C mice, (C) ICR mice. Data are expressed as accumulated total moving distances in cages (vertical bars represent standard error of the mean). *p<0.05, **p<0.01, ##p<0.01 vs day 0 (n=6~10).0

  • Fig. 4 Effect of MPTP (20 mg/kg, s.c) on total rearing frequencies until 1 hour after MPTP administration. (A) C57BL/6 mice, (B) Balb C mice, (C) ICR mice. Data are expressed as accumulated number of rearing frequencies in cages (vertical bars represent standard error of the mean). *p<0.05, **p<0.01 vs day 0 (n=6~10).

  • Fig. 5 Comparison of moving distances and total rearing frequencies among three mouse strains Effect of MPTP (20 mg/kg, s.c) on moving distances (A) and total rearing frequencies (B) until 1 hour after MPTP administration over three days. In each case, the mean results were normalized by the mean of day 0. Vertical bars represent standard error of the mean and negative SE bars have been deleted for clarity. Two-way ANOVA, *p<0.05, **p<0.01 vs ICR (n=6~10).

  • Fig. 6 Effects of MPTP (20 mg/kg, s.c) on dopamine concentration in mouse brain section. The endogenous level of dopamine in mice brain scetions ((A) striatum, (B) substantia nigra, (C) hippocampus) were measured by LC-MS/MS in three different mice species. (D) internal calibration for dopamine. Data is expressed as mean±s.e.m. *p<0.05, **p<0.01 (n=5).

  • Fig. 7 Effects of MPTP (20 mg/kg, s.c) on tetrahydrobiopterin (BH4) concentration in brain regions. The endogenous level of BH4 in mice brain regions ((A) striatum, (B) substantia nigra, (C) hippocampus) were measured by LC-MS/MS in three different mice strains. (D) Internal calibration for BH4. Data is expressed as mean±standard error of the mean (n=5).

  • Fig. 8 Effects of MPTP (20 mg/kg, s.c) on the expression of tyrosine hydroxylase in the striatum in the three strains of mice. (A) Representative immunoblots of tyrosine hydroxylase (TH) and β-actin expression. (B) Densitometry plot of TH expression immunoblots normalized to the control levels, with and without MPTP treatment (n=4). *p<0.05 and **p<0.01 vs. black bar (control group).


Cited by  1 articles

Alteration of Striatal Tetrahydrobiopterin in Iron-Induced Unilateral Model of Parkinson's Disease
Bijay Aryal, Jin-Koo Lee, Hak Rim Kim, Hyung-Gun Kim
Korean J Physiol Pharmacol. 2014;18(2):129-134.    doi: 10.4196/kjpp.2014.18.2.129.


Reference

1. Blandini F, Armentero MT. Animal models of Parkinson's disease. FEBS J. 2012; 279:1156–1166. PMID: 22251459.
Article
2. Yokoyama H, Kuroiwa H, Kasahara J, Araki T. Neuropharmacological approach against MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine)-induced mouse model of Parkinson's disease. Acta Neurobiol Exp (Wars). 2011; 71:269–280. PMID: 21731080.
3. Prediger RD, Aguiar AS Jr, Moreira EL, Matheus FC, Castro AA, Walz R, De Bem AF, Latini A, Tasca CI, Farina M, Raisman-Vozari R. The intranasal administration of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP): a new rodent model to test palliative and neuroprotective agents for Parkinson's disease. Curr Pharm Des. 2011; 17:489–507. PMID: 21375482.
4. Onyango IG. Mitochondrial dysfunction and oxidative stress in Parkinson's disease. Neurochem Res. 2008; 33:589–597. PMID: 17940895.
Article
5. Duty S, Jenner P. Animal models of Parkinson's disease: a source of novel treatments and clues to the cause of the disease. Br J Pharmacol. 2011; 164:1357–1391. PMID: 21486284.
Article
6. Giovanni A, Sieber BA, Heikkila RE, Sonsalla PK. Studies on species sensitivity to the dopaminergic neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine. Part 1: Systemic administration. J Pharmacol Exp Ther. 1994; 270:1000–1007. PMID: 7932147.
7. Giovanni A, Sonsalla PK, Heikkila RE. Studies on species sensitivity to the dopaminergic neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine. Part 2: Central administration of 1-methyl-4-phenylpyridinium. J Pharmacol Exp Ther. 1994; 270:1008–1014. PMID: 7932148.
8. Ramsay RR, Singer TP. Energy-dependent uptake of N-methyl-4-phenylpyridinium, the neurotoxic metabolite of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, by mitochondria. J Biol Chem. 1986; 261:7585–7587. PMID: 3486869.
Article
9. Nicklas WJ, Vyas I, Heikkila RE. Inhibition of NADH-linked oxidation in brain mitochondria by 1-methyl-4-phenyl-pyridine, a metabolite of the neurotoxin, 1-methyl-4-phenyl-1,2,5,6-tetrahydropyridine. Life Sci. 1985; 36:2503–2508. PMID: 2861548.
Article
10. Mizuno Y, Suzuki K, Sone N, Saitoh T. Inhibition of ATP synthesis by 1-methyl-4-phenylpyridinium ion (MPP+) in isolated mitochondria from mouse brains. Neurosci Lett. 1987; 81:204–208. PMID: 3501080.
Article
11. Di Monte D, Jewell SA, Ekström G, Sandy MS, Smith MT. 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and 1-methyl-4-phenylpyridine (MPP+) cause rapid ATP depletion in isolated hepatocytes. Biochem Biophys Res Commun. 1986; 137:310–315. PMID: 3487319.
Article
12. Volkmann J, Daniels C, Witt K. Neuropsychiatric effects of subthalamic neurostimulation in Parkinson disease. Nat Rev Neurol. 2010; 6:487–498. PMID: 20680036.
Article
13. Voon V, Fernagut PO, Wickens J, Baunez C, Rodriguez M, Pavon N, Juncos JL, Obeso JA, Bezard E. Chronic dopaminergic stimulation in Parkinson's disease: from dyskinesias to impulse control disorders. Lancet Neurol. 2009; 8:1140–1149. PMID: 19909912.
Article
14. Duch DS, Smith GK. Biosynthesis and function of tetrahydrobiopterin. J Nutr Biochem. 1991; 2:411–423.
Article
15. Schmidt TS, Alp NJ. Mechanisms for the role of tetrahydrobiopterin in endothelial function and vascular disease. Clin Sci (Lond). 2007; 113:47–63. PMID: 17555404.
Article
16. Sonsalla PK, Heikkila RE. Neurotoxic effects of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and methamphetamine in several strains of mice. Prog Neuropsychopharmacol Biol Psychiatry. 1988; 12:345–354. PMID: 3260386.
Article
17. Burns RS, Chiueh CC, Markey SP, Ebert MH, Jacobowitz DM, Kopin IJ. A primate model of parkinsonism: selective destruction of dopaminergic neurons in the pars compacta of the substantia nigra by N-methyl-4-phenyl-1,2,3,6-tetrahydropyridine. Proc Natl Acad Sci USA. 1983; 80:4546–4550. PMID: 6192438.
Article
18. Langston JW, Forno LS, Rebert CS, Irwin I. Selective nigral toxicity after systemic administration of 1-methyl-4-phenyl-1,2,5,6-tetrahydropyrine (MPTP) in the squirrel monkey. Brain Res. 1984; 292:390–394. PMID: 6607092.
19. Antzoulatos E, Jakowec MW, Petzinger GM, Wood RI. Sex differences in motor behavior in the MPTP mouse model of Parkinson's disease. Pharmacol Biochem Behav. 2010; 95:466–472. PMID: 20347863.
Article
20. Kim HR, Kim TH, Hong SH, Kim HG. Direct detection of tetrahydrobiopterin (BH4) and dopamine in rat brain using liquid chromatography coupled electrospray tandem mass spectrometry. Biochem Biophys Res Commun. 2012; 419:632–637. PMID: 22382017.
Article
21. Kim HR, Graceffa P, Ferron F, Gallant C, Boczkowska M, Dominguez R, Morgan KG. Actin polymerization in differentiated vascular smooth muscle cells requires vasodilator-stimulated phosphoprotein. Am J Physiol Cell Physiol. 2010; 298:C559–C571. PMID: 20018948.
Article
22. Schwarting RK, Sedelis M, Hofele K, Auburger GW, Huston JP. Strain-dependent recovery of open-field behavior and striatal dopamine deficiency in the mouse MPTP model of Parkinson's disease. Neurotox Res. 1999; 1:41–56. PMID: 12835113.
Article
23. Choi HJ, Lee SY, Cho Y, No H, Kim SW, Hwang O. Tetrahydrobiopterin causes mitochondrial dysfunction in dopaminergic cells: implications for Parkinson's disease. Neurochem Int. 2006; 48:255–262. PMID: 16343695.
Article
24. Ichinose H, Nomura T, Sumi-Ichinose C. Metabolism of tetrahydrobiopterin: its relevance in monoaminergic neurons and neurological disorders. Chem Rec. 2008; 8:378–385. PMID: 19107867.
Article
25. Foxton RH, Land JM, Heales SJ. Tetrahydrobiopterin availability in Parkinson's and Alzheimer's disease potential pathogenic mechanisms. Neurochem Res. 2007; 32:751–756. PMID: 17191137.
Article
26. Takazawa C, Fujimoto K, Homma D, Sumi-Ichinose C, Nomura T, Ichinose H, Katoh S. A brain-specific decrease of the tyrosine hydroxylase protein in sepiapterin reductase-null mice--as a mouse model for Parkinson's disease. Biochem Biophys Res Commun. 2008; 367:787–792. PMID: 18201550.
Article
27. Sundström E, Strömberg I, Tsutsumi T, Olson L, Jonsson G. Studies on the effect of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) on central catecholamine neurons in C57BL/6 mice. Comparison with three other strains of mice. Brain Res. 1987; 405:26–38. PMID: 2882814.
28. Obeso JA, Rodriguez-Oroz MC, Goetz CG, Marin C, Kordower JH, Rodriguez M, Hirsch EC, Farrer M, Schapira AH, Halliday G. Missing pieces in the Parkinson's disease puzzle. Nat Med. 2010; 16:653–661. PMID: 20495568.
Article
29. Nagatsu T, Sawada M. Molecular mechanism of the relation of monoamine oxidase B and its inhibitors to Parkinson's disease: possible implications of glial cells. J Neural Transm Suppl. 2006; (71):53–65. PMID: 17447416.
Article
30. Francardo V, Recchia A, Popovic N, Andersson D, Nissbrandt H, Cenci MA. Impact of the lesion procedure on the profiles of motor impairment and molecular responsiveness to L-DOPA in the 6-hydroxydopamine mouse model of Parkinson's disease. Neurobiol Dis. 2011; 42:327–340. PMID: 21310234.
Article
31. McMillan PJ, White SS, Franklin A, Greenup JL, Leverenz JB, Raskind MA, Szot P. Differential response of the central noradrenergic nervous system to the loss of locus coeruleus neurons in Parkinson's disease and Alzheimer's disease. Brain Res. 2011; 1373:240–252. PMID: 21147074.
Article
32. Kitada T, Tomlinson JJ, Ao HS, Grimes DA, Schlossmacher MG. Considerations regarding the etiology and future treatment of autosomal recessive versus idiopathic Parkinson disease. Curr Treat Options Neurol. 2012; 14:230–240. PMID: 22547255.
Article
33. Sedelis M, Hofele K, Auburger GW, Morgan S, Huston JP, Schwarting RK. MPTP susceptibility in the mouse: behavioral, neurochemical, and histological analysis of gender and strain differences. Behav Genet. 2000; 30:171–182. PMID: 11105391.
Full Text Links
  • KJPP
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr