Publication Cover
Neurological Research
A Journal of Progress in Neurosurgery, Neurology and Neurosciences
Volume 39, 2017 - Issue 5
260
Views
1
CrossRef citations to date
0
Altmetric
Original Research Paper

Effects of eldepryl on glial cell proliferation and activation in the substantia nigra and striatum in a rat model of Parkinson’s disease

, , , , , , , & show all
Pages 459-467 | Received 10 Sep 2016, Accepted 17 Feb 2017, Published online: 09 Mar 2017

References

  • Keane H, Ryan BJ, Jackson B, et al. Protein-protein interaction networks identify targets which rescue the MPP(+) cellular model of Parkinson’s disease. Sci Rep. 2015;5:17004.10.1038/srep17004
  • Lee HJ, Han J, Jang Y, et al. Docosahexaenoic acid prevents paraquat-induced reactive oxygen species production in dopaminergic neurons via enhancement of glutathione homeostasis. Biochem Biophys Res Commun. 2015;457:95–100.10.1016/j.bbrc.2014.12.085
  • Xu W, Tan L, Yu JT. Link between the SNCA gene and parkinsonism. Neurobiol Aging. 2015;36:1505–1518.10.1016/j.neurobiolaging.2014.10.042
  • Han JY, Kim JS, Son JH. Mitochondrial homeostasis molecules: regulation by a trio of recessive Parkinson’s disease genes. Exp Neurobiol. 2014;23:345–351.10.5607/en.2014.23.4.345
  • Pessoa Rocha N, Reis HJ, Vanden Berghe P, et al. Depression and cognitive impairment in Parkinson’s disease: a role for inflammation and immunomodulation? Neuroimmunomodulation. 2014;21:88–94.
  • González H, Elgueta D, Montoya A, et al. Neuroimmune regulation of microglial activity involved in neuroinflammation and neurodegenerative diseases. J Neuroimmunol. 2014;274:1–13.10.1016/j.jneuroim.2014.07.012
  • Hirsch EC, Vyas S, Hunot S. Neuroinflammation in Parkinson’s disease. Parkinsonism Relat Disord. 2012;18:S210–S212.10.1016/S1353-8020(11)70065-7
  • Long-Smith CM, Sullivan AM, Nolan YM. The influence of microglia on the pathogenesis of Parkinson’s disease. Prog Neurobiol. 2009;89:277–287.10.1016/j.pneurobio.2009.08.001
  • Doorn KJ, Moors T, Drukarch B, et al. Microglial phenotypes and toll-like receptor 2 in the substantia nigra and hippocampus of incidental Lewy body disease cases and Parkinson’s disease patients. Acta Neuropathol Commun. 2014;2:90.
  • Doorn KJ, Lucassen PJ, Boddeke HW, et al. Emerging roles of microglial activation and non-motor symptoms in Parkinson’s disease. Prog Neurobiol. 2012;98:222–238.10.1016/j.pneurobio.2012.06.005
  • Salemme A, Togna AR, Mastrofrancesco A, et al. Anti-inflammatory effects and antioxidant activity of dihydroasparagusic acid in lipopolysaccharide-activated microglial cells. Brain Res Bull. 2016;120:151–158.10.1016/j.brainresbull.2015.11.014
  • Jha MK, Suk K. Glia-based biomarkers and their functional role in the CNS. Expert Rev Proteomics. 2013;10:43–63.10.1586/epr.12.70
  • Weinreb O, Amit T, Sagi Y, et al. Genomic and proteomic study to survey the mechanism of action of the anti-Parkinson’s disease drug, rasagiline compared with selegiline, in the rat midbrain. J Neural Transm. 2009;116:1457–1472.10.1007/s00702-009-0225-x
  • Kim M, Choi SY, Lee P, et al. Neochlorogenic acid inhibits lipopolysaccharide-induced activation and pro-inflammatory responses in BV2 microglial cells. Neurochem Res. 2015;40:1792–1798.10.1007/s11064-015-1659-1
  • Batassini C, Broetto N, Tortorelli LS, et al. Striatal Injury with 6-OHDA transiently increases cerebrospinal GFAP and S100B. Neural Plast. 2015;2015: 387028.
  • Devos D, Lebouvier T, Lardeux B, et al. Colonic inflammation in Parkinson’s disease. Neurobiol Dis. 2013;50:42–48.10.1016/j.nbd.2012.09.007
  • Mika J, Wawrzczak-Bargiela A, Osikowicz M, et al. Attenuation of morphine tolerance by minocycline and pentoxifylline in naive and neuropathic mice. Brain Behav Immun. 2009;23:75–84.10.1016/j.bbi.2008.07.005
  • Maolood N, Meister B. Protein components of the blood-brain barrier (BBB) in the brainstem area postrema-nucleus tractus solitarius region. J Chem Neuroanat. 2009;37:182–195.10.1016/j.jchemneu.2008.12.007
  • Zhao Q, Cai D, Bai Y. Selegiline rescues gait deficits and the loss of dopaminergic neurons in a subacute MPTP mouse model of Parkinson’s disease. Int J Mol Med. 2013;32:883–891.
  • Robottom BJ. Efficacy, safety, and patient preference of monoamine oxidase B inhibitors in the treatment of Parkinson’s disease. Patient Prefer Adherence. 2011;5:57–64.10.2147/PPA
  • Fabbrini G, Abbruzzese G, Marconi S, et al. Selegiline: a reappraisal of its role in Parkinson disease. Clin Neuropharmacol. 2012;35:134–140.10.1097/WNF.0b013e318255838b
  • Peretz C, Segev H, Rozani V, et al. Comparison of selegiline and rasagiline therapies in Parkinson Disease: a real-life study. Clin Neuropharmacol. 2016;39(5):227–231.10.1097/WNF.0000000000000167
  • Liu B, Sun J, Zhang J, et al. Autophagy-related protein expression in the substantia nigra and eldepryl intervention in rat models of Parkinson’s disease. Brain Res. 2015;1625:180–188.10.1016/j.brainres.2015.08.033
  • Zhao Q, Cai D, Bai Y. Selegiline rescues gait deficits and the loss of dopaminergic neurons in a subacute MPTP mouse model of Parkinson’s disease. Int J Mol Med. 2013;32:883–891.
  • Weinreb O, Amit T, Sagi Y, et al. Genomic and proteomic study to survey the mechanism of action of the anti-Parkinson’s disease drug, rasagiline compared with selegiline, in the rat midbrain. J Neural Transm. 2009;116:1457–1472.10.1007/s00702-009-0225-x
  • Feng J. Microtubule: a common target for Parkin and Parkinson’s disease toxins. Neuroscientist. 2006;12:469–476.10.1177/1073858406293853
  • Sherer TB, Betarbet R, Kim JH, et al. Selective microglial activation in the rat rotenone model of Parkinson’s disease. Neurosci Lett. 2003;341:87–90.10.1016/S0304-3940(03)00172-1
  • Grieb B, von Nicolai C, Engler G, et al. Decomposition of abnormal free locomotor behavior in a rat model of Parkinson’s disease. Front Syst Neurosci. 2013;7:95.
  • Bartolomei F, Massacrier A, Rey M, et al. Effect of gamma knife radiosurgery on rat brain sodium channel subunit mRNA expression. Stereotact Funct Neurosurg. 1998;70:237–242.10.1159/000056427
  • Sánchez-González A, Mendieta L, Palafox V, et al. The restorative effect of intramuscular injection of tetanus toxin C-fragment in hemiparkinsonian rats. Neurosci Res. 2014;84:1–9.10.1016/j.neures.2014.04.008
  • Carriere CH, Kang NH, Niles LP. Neuroprotection by valproic acid in an intrastriatal rotenone model of Parkinson’s disease. Neuroscience. 2014;267:114–121.10.1016/j.neuroscience.2014.02.028
  • Li C, Chen X, Zhang N, et al. Gastrodin inhibits neuroinflammation in rotenone-induced Parkinson’s disease model rats. Neural Regen Res. 2012;7:325–331.
  • Dexter DT, Jenner P. Parkinson disease: from pathology to molecular disease mechanisms. Free Radic Biol Med. 2013;62:132–144.10.1016/j.freeradbiomed.2013.01.018
  • Wang Q, Liu Y, Zhou J. Neuroinflammation in Parkinson’s disease and its potential as therapeutic target. Transl Neurodegener. 2015;4:19.10.1186/s40035-015-0042-0
  • Tripathy D, Chakraborty J, Mohanakumar KP. Antagonistic pleiotropic effects of nitric oxide in the pathophysiology of Parkinson’s disease. Free Radic Res. 2015;49:1129–1139.10.3109/10715762.2015.1045505
  • Niranjan R. The role of inflammatory and oxidative stress mechanisms in the pathogenesis of Parkinson’s disease: focus on astrocytes. Mol Neurobiol. 2014;49:28–38.10.1007/s12035-013-8483-x
  • Saijo K, Winner B, Carson CT, et al. A Nurr1/CoREST pathway in microglia and astrocytes protects dopaminergic neurons from inflammation-induced death. Cell. 2009;137:47–59.10.1016/j.cell.2009.01.038
  • Kim BW, Koppula S, Hong SS, et al. Regulation of microglia activity by glaucocalyxin-A: attenuation of lipopolysaccharide-stimulated neuroinflammation through NF-κB and p38 MAPK signaling pathways. PLoS One. 2013;8:e55792.10.1371/journal.pone.0055792
  • Kim SU, de Vellis J. Microglia in health and disease. J Neurosci Res. 2005;81:302–313.10.1002/(ISSN)1097-4547
  • Herrero MT, Estrada C, Maatouk L, et al. Inflammation in Parkinson’s disease: role of glucocorticoids. Front Neuroanat. 2015;9:32.
  • Tufekci KU, Genc S, Genc K. The endotoxin-induced neuroinflammation model of Parkinson’s disease. Parkinsons Dis. 2011;2011: 487450.
  • Amor S, Puentes F, Baker D, et al. Inflammation in neurodegenerative diseases. Immunology. 2010;129:154–169.
  • Shukla V, Mishra SK, Pant HC. Oxidative stress in neurodegeneration. Adv Pharmacol Sci. 2011;2011: 572634.
  • Dexter DT, Jenner P. Parkinson Disease: from pathology to molecular disease mechanisms. Free Radical Bio Med. 2013;62:132–144.10.1016/j.freeradbiomed.2013.01.018
  • González H, Elgueta D, Montoya A, et al. Neuroimmune regulation of microglial activity involved in neuroinflammation and neurodegenerative diseases. J Neuroimmunol. 2014;274:1–13.10.1016/j.jneuroim.2014.07.012
  • Gao HM, Hong JS. Why neurodegenerative diseases are progressive: uncontrolled inflammation drives disease progression. Trends Immunol. 2008;29:357–365.10.1016/j.it.2008.05.002
  • Brochard V, Combadière B, Prigent A, et al. Infiltration of CD4+ lymphocytes into the brain contributes to neurodegeneration in a mouse model of Parkinson disease. J Clin Invest. 2009;119:182–192.
  • Harms AS, Cao S, Rowse AL, et al. MHCII is required for α-synuclein-induced activation of microglia, CD4 T cell proliferation, and dopaminergic neurodegeneration. J Neurosci. 2013;33:9592–9600.10.1523/JNEUROSCI.5610-12.2013
  • Noelker C, Morel L, Lescot T, et al. Toll like receptor 4 mediates cell death in a mouse MPTP model of Parkinson disease. Sci Rep. 2013;3:1393.
  • Connolly BS, Lang AE. Pharmacological treatment of Parkinson disease: a review. JAMA. 2014;311:1670–1683.10.1001/jama.2014.3654
  • Finberg JP. Update on the pharmacology of selective inhibitors of MAO-A and MAO-B: focus on modulation of CNS monoamine neurotransmitter release. Pharmacol Ther. 2014;143:133–152.10.1016/j.pharmthera.2014.02.010
  • Lieu CA, Chinta SJ, Rane A, et al. Age-related behavioral phenotype of an astrocytic monoamine oxidase-B transgenic mouse model of Parkinson’s disease. PLoS One. 2013;8:e54200.10.1371/journal.pone.0054200
  • Chen JJ, Wilkinson JR. The monoamine oxidase type B inhibitor rasagiline in the treatment of Parkinson disease: is tyramine a challenge? J Clin Pharmacol. 2012;52:620–628.10.1177/0091270011406279
  • Mattsson C, Svensson P, Sonesson C. A novel series of 6-substituted 3-(pyrrolidin-1-ylmethyl)chromen-2-ones as selective monoamine oxidase (MAO) A inhibitors. Eur J Med Chem. 2014;73:177–186.10.1016/j.ejmech.2013.11.035
  • Schwartz TL. A neuroscientific update on monoamine oxidase and its inhibitors. CNS Spectr. 2013;18:25–32; quiz 33.
  • Boppana K, Dubey PK, Jagarlapudi SA, et al. Knowledge based identification of MAO-B selective inhibitors using pharmacophore and structure based virtual screening models. Eur J Med Chem. 2009;44:3584–3590.10.1016/j.ejmech.2009.02.031
  • Naoi M, Maruyama W. Monoamine oxidase inhibitors as neuroprotective agents in age-dependent neurodegenerative disorders. Curr Pharm Des. 2010;16:2799–2817.10.2174/138161210793176527
  • Youdim MB, Bakhle YS. Monoamine oxidase: isoforms and inhibitors in Parkinson’s disease and depressive illness. Br J Pharmacol. 2006;147:S287–S296.
  • LeWitt PA. Clinical trials of neuroprotection for Parkinson’s disease. Neurology. 2004;63:S23–S31.10.1212/WNL.63.7_suppl_2.S23
  • Robottom BJ. Efficacy, safety, and patient preference of monoamine oxidase B inhibitors in the treatment of Parkinson’s disease. Patient Prefer Adherence. 2011;5:57–64.10.2147/PPA

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.