Skip to main content
Top
Published in: Journal of Neuroinflammation 1/2016

Open Access 01-12-2016 | Research

Long-term trihexyphenidyl exposure alters neuroimmune response and inflammation in aging rat: relevance to age and Alzheimer’s disease

Authors: Yuqi Huang, Zhe Zhao, Xiaoli Wei, Yong Zheng, Jianqiang Yu, Jianquan Zheng, Liyun Wang

Published in: Journal of Neuroinflammation | Issue 1/2016

Login to get access

Abstract

Background

Clinical studies have shown an association between long-term anticholinergic (AC) drug exposure and Alzheimer’s disease (AD) pathogenesis, which has been primarily investigated in Parkinson’s disease (PD). However, long-term AC exposure as a risk factor for developing neurodegenerative disorders and the exact mechanisms and potential for disease progression remain unclear. Here, we have addressed the issue using trihexyphenidyl (THP), a commonly used AC drug in PD patients, to determine if THP can accelerate AD-like neurodegenerative progression and study potential mechanisms involved.

Methods

Male Sprague-Dawley rats (SD) were intraperitoneally injected with THP (0.3 and 1.0 mg/kg) or normal saline (NS) for 7 months. Alterations in cognitive and behavioral performance were assessed using the Morris water maze (MWM) and open field tests. After behavior tests, whole genome oligo microarrays, quantitative real-time reverse transcription-polymerase chain reaction (qRT-PCR), immunohistochemistry, and immunofluorescence-confocal were used to investigate the global mechanisms underlying THP-induced neuropathology with aging.

Results

Compared with NS controls, the MWM test results showed that THP-treated rats exhibited significantly extended mean latencies during the initial 3 months of testing; however, this behavioral deficit was restored between the fourth and sixth month of MWM testing. The same tendencies were confirmed by MWM probe and open field tests. Gene microarray analysis identified 68 (47 %) upregulated and 176 (53 %) downregulated genes in the “THP-aging” vs. “NS-aging” group. The most significant populations of genes downregulated by THP were the immune response-, antigen processing and presentation-, and major histocompatibility complex (MHC)-related genes, as validated by qRT-PCR. The decreased expression of MHC class I in THP-treated aging brains was confirmed by confocal analysis. Notably, long-term THP treatment primed hippocampal and cortical microglia to undergo an inflammatory phenotypic switch, causing microgliosis and microglia activation, which were positively accompanied by pathological misfolded tau lesions.

Conclusions

Our findings suggest that immune response and neuroinflammation represent a pivotal mechanism in THP-induced AD-like neuropathology processes with long-term exposure to AC drugs.
Appendix
Available only for authorised users
Literature
1.
go back to reference Flicker C, Serby M, Ferris SH. Scopolamine effects on memory, language, visuospatial praxis and psychomotor speed. Psychopharmacology (Berl). 1990;100:243–50.CrossRef Flicker C, Serby M, Ferris SH. Scopolamine effects on memory, language, visuospatial praxis and psychomotor speed. Psychopharmacology (Berl). 1990;100:243–50.CrossRef
2.
go back to reference Ancelin ML, Artero S, Portet F, Dupuy AM, Touchon J, Rithie K. Nondegenerative mild cognitive impairment in elderly people and use of anticholinergic drugs: longitudinal cohort study. BMJ. 2006;332:455–9.CrossRefPubMedPubMedCentral Ancelin ML, Artero S, Portet F, Dupuy AM, Touchon J, Rithie K. Nondegenerative mild cognitive impairment in elderly people and use of anticholinergic drugs: longitudinal cohort study. BMJ. 2006;332:455–9.CrossRefPubMedPubMedCentral
3.
go back to reference Boustani M, Campbell NL, Munger S, Maidment I, Fox C. Impact of anticholinergics on the aging brain: a review and practical application. Aging Health. 2008;4:311–20.CrossRef Boustani M, Campbell NL, Munger S, Maidment I, Fox C. Impact of anticholinergics on the aging brain: a review and practical application. Aging Health. 2008;4:311–20.CrossRef
4.
go back to reference Perry EK, Kilford L, Lees AJ, Burn DJ, Perry RH. Increased Alzheimer pathology in Parkinson’s disease related to antimuscarinic drugs. Ann Neurol. 2003;54:235–8.CrossRefPubMed Perry EK, Kilford L, Lees AJ, Burn DJ, Perry RH. Increased Alzheimer pathology in Parkinson’s disease related to antimuscarinic drugs. Ann Neurol. 2003;54:235–8.CrossRefPubMed
5.
go back to reference Kajimura N, Mizuki Y, Kai S, Suetsugi M, Yamada M, Okuma T. Memory and cognitive impairments in a case of long-term trihexyphenidyl abuse. Pharmacopsychiatry. 1993;26:59–62.CrossRefPubMed Kajimura N, Mizuki Y, Kai S, Suetsugi M, Yamada M, Okuma T. Memory and cognitive impairments in a case of long-term trihexyphenidyl abuse. Pharmacopsychiatry. 1993;26:59–62.CrossRefPubMed
6.
go back to reference Takahashi S, Tohgi H, Yonezawa H, Obara S, Yamazaki E. The effect of trihexyphenidyl, an anticholinergic agent, on regional cerebral blood flow and oxygen metabolism in patients with Parkinson’s disease. J Neurol Sci. 1999;167:56–61.CrossRefPubMed Takahashi S, Tohgi H, Yonezawa H, Obara S, Yamazaki E. The effect of trihexyphenidyl, an anticholinergic agent, on regional cerebral blood flow and oxygen metabolism in patients with Parkinson’s disease. J Neurol Sci. 1999;167:56–61.CrossRefPubMed
7.
go back to reference Welt T, Kulic L, Hoey SE, McAfoose J, Späni C, Chadha AS, Fisher A, Nitsch RM. Acute effects of muscarinic M1 receptor modulation on AβPP metabolism and amyloid-β levels in vivo: a microdialysis study. J Alzheimers Dis. 2015;46:971–82.CrossRefPubMed Welt T, Kulic L, Hoey SE, McAfoose J, Späni C, Chadha AS, Fisher A, Nitsch RM. Acute effects of muscarinic M1 receptor modulation on AβPP metabolism and amyloid-β levels in vivo: a microdialysis study. J Alzheimers Dis. 2015;46:971–82.CrossRefPubMed
8.
go back to reference Haring R, Fisher A, Marciano D, Pittel Z, Kloog Y, Zuckerman A, et al. Mitogen-activated protein kinase-dependent and protein kinase C-dependent pathways link the m1 muscarinic receptor to b-amyloid protein secretion. J Neurochem. 1998;71:2094–103.CrossRefPubMed Haring R, Fisher A, Marciano D, Pittel Z, Kloog Y, Zuckerman A, et al. Mitogen-activated protein kinase-dependent and protein kinase C-dependent pathways link the m1 muscarinic receptor to b-amyloid protein secretion. J Neurochem. 1998;71:2094–103.CrossRefPubMed
9.
go back to reference Tanda G, Ebbs AL, Kopajtic TA, Elias LM, Campbell BL, Newman AH, Katz JL. Effects of muscarinic M1 receptor blockade on cocaine-induced elevations of brain dopamine levels and locomotor behavior in rats. J Pharmacol Exp Ther. 2007;321:334–44.CrossRefPubMed Tanda G, Ebbs AL, Kopajtic TA, Elias LM, Campbell BL, Newman AH, Katz JL. Effects of muscarinic M1 receptor blockade on cocaine-induced elevations of brain dopamine levels and locomotor behavior in rats. J Pharmacol Exp Ther. 2007;321:334–44.CrossRefPubMed
10.
go back to reference Bédard MA, Pillon B, Dubois B, Duchesne N, Masson H, Agid Y. Acute and long-term administration of anticholinergics in Parkinson's disease: specific effects on the subcortico-frontal syndrome. Brain Cogn. 1999;40:289–313.CrossRefPubMed Bédard MA, Pillon B, Dubois B, Duchesne N, Masson H, Agid Y. Acute and long-term administration of anticholinergics in Parkinson's disease: specific effects on the subcortico-frontal syndrome. Brain Cogn. 1999;40:289–313.CrossRefPubMed
11.
go back to reference Nelson RL, Guo Z, Halagappa VM, Pearson M, Gray AJ, et al. Prophylactic treatment with paroxetine ameliorates behavioral deficits and retards the development of amyloid and tau pathologies in 36TgAD mice. Exp Neurol. 2007;205:166–76.CrossRefPubMedPubMedCentral Nelson RL, Guo Z, Halagappa VM, Pearson M, Gray AJ, et al. Prophylactic treatment with paroxetine ameliorates behavioral deficits and retards the development of amyloid and tau pathologies in 36TgAD mice. Exp Neurol. 2007;205:166–76.CrossRefPubMedPubMedCentral
12.
go back to reference Boger HA, Middaugh LD, Huang P, Zaman V, Smith AC, Hoffer BJ, Tomac AC, Granholm AC. A partial GDNF depletion leads to earlier age-related deterioration of motor function and tyrosine hydroxylase expression in the substantia nigra. Exp Neurol. 2006;202:336–47.CrossRefPubMed Boger HA, Middaugh LD, Huang P, Zaman V, Smith AC, Hoffer BJ, Tomac AC, Granholm AC. A partial GDNF depletion leads to earlier age-related deterioration of motor function and tyrosine hydroxylase expression in the substantia nigra. Exp Neurol. 2006;202:336–47.CrossRefPubMed
13.
go back to reference Martin B, Pearson M, Brenneman R, Golden E, Wood W, et al. Gonadal transcriptome alterations in response to dietary energy intake: sensing the reproductive environment. PLoS One. 2009. doi:10.1371/journal.pone.0004146. Martin B, Pearson M, Brenneman R, Golden E, Wood W, et al. Gonadal transcriptome alterations in response to dietary energy intake: sensing the reproductive environment. PLoS One. 2009. doi:10.​1371/​journal.​pone.​0004146.
14.
go back to reference Ding L et al. Human four-and-a-half LIM family members suppress tumor cell growth through a TGF-β-like signaling pathway. J Clin Invest. 2009;119:349–61.PubMedPubMedCentral Ding L et al. Human four-and-a-half LIM family members suppress tumor cell growth through a TGF-β-like signaling pathway. J Clin Invest. 2009;119:349–61.PubMedPubMedCentral
15.
go back to reference Krstic D, Madhusudan A, Doehner J, Vogel P, Notter T, Imhof C, Manalastas A, Hilfiker M, Pfister S, Schwerdel C, Riether C, Meyer U, Knuesel I. Systemic immune challenges trigger and drive Alzheimer-like neuropathology in mice. J Neuroinflammation. 2012;2(I):9–151. Krstic D, Madhusudan A, Doehner J, Vogel P, Notter T, Imhof C, Manalastas A, Hilfiker M, Pfister S, Schwerdel C, Riether C, Meyer U, Knuesel I. Systemic immune challenges trigger and drive Alzheimer-like neuropathology in mice. J Neuroinflammation. 2012;2(I):9–151.
16.
go back to reference Gallyas F. Silver staining of Alzheimer’s neurofibrillary changes by means of physical development. Acta Morphol Acad Sci Hung. 1971;19:1–8.PubMed Gallyas F. Silver staining of Alzheimer’s neurofibrillary changes by means of physical development. Acta Morphol Acad Sci Hung. 1971;19:1–8.PubMed
17.
go back to reference Moser MB, Moser EI. Distributed encoding and retrieval of spatial memory in the hippocampus. J Neurosci. 1998;18:7535–42.PubMed Moser MB, Moser EI. Distributed encoding and retrieval of spatial memory in the hippocampus. J Neurosci. 1998;18:7535–42.PubMed
18.
go back to reference Terry Jr AV. Spatial navigation (water maze) tasks. In: Buccafusco JJ, editor. Methods of behavior analysis in neuroscience. Boca Raton (FL): CRC Press/Taylor & Francis; 2009. Chapter 13. Terry Jr AV. Spatial navigation (water maze) tasks. In: Buccafusco JJ, editor. Methods of behavior analysis in neuroscience. Boca Raton (FL): CRC Press/Taylor & Francis; 2009. Chapter 13.
19.
go back to reference Klinkenberg I, Blokland A. The validity of scopolamine as a pharmacological model for cognitive impairment: a review of animal behavioral studies. Neurosci Biobehav Rev. 2010;34:1307–50.CrossRefPubMed Klinkenberg I, Blokland A. The validity of scopolamine as a pharmacological model for cognitive impairment: a review of animal behavioral studies. Neurosci Biobehav Rev. 2010;34:1307–50.CrossRefPubMed
20.
go back to reference Blokland A. Acetylcholine: a neurotransmitter for learning and memory? Brain Res Rev. 1995;21:285–300.CrossRefPubMed Blokland A. Acetylcholine: a neurotransmitter for learning and memory? Brain Res Rev. 1995;21:285–300.CrossRefPubMed
21.
go back to reference Stozicka Z, Zilka N, Novak P, Kovacech B, Bugos O, Novak M. Genetic background modifies neurodegeneration and neuroinflammation driven by misfolded human tau protein in rat model of tauopathy: implication for immunomodulatory approach to Alzheimer’s disease. J Neuroinflammation. 2010. doi:10.1186/1742-2094-7-64.PubMedPubMedCentral Stozicka Z, Zilka N, Novak P, Kovacech B, Bugos O, Novak M. Genetic background modifies neurodegeneration and neuroinflammation driven by misfolded human tau protein in rat model of tauopathy: implication for immunomodulatory approach to Alzheimer’s disease. J Neuroinflammation. 2010. doi:10.​1186/​1742-2094-7-64.PubMedPubMedCentral
22.
go back to reference Perry VH, Teeling J. Microglia and macrophages of the central nervous system: the contribution of microglia priming and systemic inflammation to chronic neurodegeneration. Semin Immunopathol. 2013;35:601–12.CrossRefPubMedPubMedCentral Perry VH, Teeling J. Microglia and macrophages of the central nervous system: the contribution of microglia priming and systemic inflammation to chronic neurodegeneration. Semin Immunopathol. 2013;35:601–12.CrossRefPubMedPubMedCentral
23.
go back to reference Fox C, Livingston G, Maidment ID, Coulton S, Smithard DG, Boustani M, Katona C. The impact of anticholinergic burden in Alzheimer's dementia-the LASER-AD study. Age Ageing. 2011;40:730–5. Fox C, Livingston G, Maidment ID, Coulton S, Smithard DG, Boustani M, Katona C. The impact of anticholinergic burden in Alzheimer's dementia-the LASER-AD study. Age Ageing. 2011;40:730–5.
24.
go back to reference Cai X, Campbell N, Khan B, Callahan C, Boustani M. Long-term anticholinergic use and the aging brain. Alzheimers Dement. 2013;9:377–85.CrossRefPubMed Cai X, Campbell N, Khan B, Callahan C, Boustani M. Long-term anticholinergic use and the aging brain. Alzheimers Dement. 2013;9:377–85.CrossRefPubMed
26.
go back to reference Zilka N, Kazmerova Z, Jadhav S, Neradil P, Madari A, Obetkova D, Bugos O, Novak M. Who fans the flames of Alzheimer’s disease brains? Misfolded tau on the crossroad of neurodegenerative and inflammatory pathways. J Neuroinflammation. 2012;9:47.CrossRefPubMedPubMedCentral Zilka N, Kazmerova Z, Jadhav S, Neradil P, Madari A, Obetkova D, Bugos O, Novak M. Who fans the flames of Alzheimer’s disease brains? Misfolded tau on the crossroad of neurodegenerative and inflammatory pathways. J Neuroinflammation. 2012;9:47.CrossRefPubMedPubMedCentral
27.
go back to reference Casserly I, Topol E. Convergence of atherosclerosis and Alzheimer’s disease: inflammation, cholesterol, and misfolded proteins. Lancet. 2004;363:1139–46.CrossRefPubMed Casserly I, Topol E. Convergence of atherosclerosis and Alzheimer’s disease: inflammation, cholesterol, and misfolded proteins. Lancet. 2004;363:1139–46.CrossRefPubMed
28.
go back to reference Ryman D, Lamb BT. Genetic and environmental modifiers of Alzheimer’s disease phenotypes in the mouse. Curr Alzheimer Res. 2006;3:465–73.CrossRefPubMed Ryman D, Lamb BT. Genetic and environmental modifiers of Alzheimer’s disease phenotypes in the mouse. Curr Alzheimer Res. 2006;3:465–73.CrossRefPubMed
29.
go back to reference Stozicka Z, Zilka N, Novak M. Risk and protective factors for sporadic Alzheimer’s disease. Acta Virol. 2007;51:205–22.PubMed Stozicka Z, Zilka N, Novak M. Risk and protective factors for sporadic Alzheimer’s disease. Acta Virol. 2007;51:205–22.PubMed
Metadata
Title
Long-term trihexyphenidyl exposure alters neuroimmune response and inflammation in aging rat: relevance to age and Alzheimer’s disease
Authors
Yuqi Huang
Zhe Zhao
Xiaoli Wei
Yong Zheng
Jianqiang Yu
Jianquan Zheng
Liyun Wang
Publication date
01-12-2016
Publisher
BioMed Central
Published in
Journal of Neuroinflammation / Issue 1/2016
Electronic ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-016-0640-5

Other articles of this Issue 1/2016

Journal of Neuroinflammation 1/2016 Go to the issue