Chronic stress enhances microglia activation and exacerbates death of nigral dopaminergic neurons under conditions of inflammation

Rocío M de Pablos, Antonio J Herrera, Ana M Espinosa-Oliva, Manuel Sarmiento, Mario F Muñoz, Alberto Machado, José L Venero, Rocío M de Pablos, Antonio J Herrera, Ana M Espinosa-Oliva, Manuel Sarmiento, Mario F Muñoz, Alberto Machado, José L Venero

Abstract

Background: Parkinson's disease is an irreversible neurodegenerative disease linked to progressive movement disorders and is accompanied by an inflammatory reaction that is believed to contribute to its pathogenesis. Since sensitivity to inflammation is not the same in all brain structures, the aim of this work was to test whether physiological conditions as stress could enhance susceptibility to inflammation in the substantia nigra, where death of dopaminergic neurons takes place in Parkinson's disease.

Methods: To achieve our aim, we induced an inflammatory process in nonstressed and stressed rats (subject to a chronic variate stress) by a single intranigral injection of lipopolysaccharide, a potent proinflammogen. The effect of this treatment was evaluated on inflammatory markers as well as on neuronal and glial populations.

Results: Data showed a synergistic effect between inflammation and stress, thus resulting in higher microglial activation and expression of proinflammatory markers. More important, the higher inflammatory response seen in stressed animals was associated with a higher rate of death of dopaminergic neurons in the substantia nigra, the most characteristic feature seen in Parkinson's disease. This effect was dependent on glucocorticoids.

Conclusions: Our data demonstrate that stress sensitises midbrain microglia to further inflammatory stimulus. This suggests that stress may be an important risk factor in the degenerative processes and symptoms of Parkinson's disease.

Figures

Figure 1
Figure 1
Experimental groups and treatments. Intranigral injections of vehicle (Veh) or lipopolysaccharide (LPS) were given at day 1. In the stressed groups (S, SL and SLR), intranigral injections were carried out after the application of the first stressor (10 minutes of forced swimming). In the SLR group, RU486 (mifepristone (11β-[p-(dimethylamino)phenyl]-17β-hydroxy-17-(1-propynyl)estra-4,9-dien-3-one)) was injected subcutaneously every day 1 hour before the stressor. V, vehicle; S, stress; L, lipopolysaccharide; SL, lipopolysaccharide injected into stressed animals; SLR, lipopolysaccharide injected into stressed animals treated with RU486.
Figure 2
Figure 2
Validation of the stress model. (A) Body weight gain (in grams). *P < 0.05. (B) Adrenal gland weight (in milligrams; bars) and ratio between adrenal gland weight and body weight gain (scatterplot and line). **P < 0.01, #P < 0.01 (for adrenal gland weight/body weight gain ratio). (C) Serum corticosterone (percentage of control animals). *P < 0.01 compared with control, aP < 0.01 compared with previous time point (S1d to S10d indicates days subjected to variate stress). Statistical significance was calculated by using Student’s t-test to compare data before (C) and after 10 days (S10d) of variate stress. Data were derived from one-way analysis of variance followed by the least significant difference (LSD) post hoc test for multiple range comparisons.
Figure 3
Figure 3
Effect of chronic stress on the lipopolysaccharide-induced activation of microglia in the ventral mesencephalon. Midbrain microglia were evaluated by immunohistochemistry with Iba-1 (left and middle columns) and OX-6 antibodies (right column) in vehicle-injected animals (A) through (F) and lipopolysaccharide (LPS)-injected animals (G) through (L) under nonstressed conditions ((A) through C) and (G) through (I)) and stressed conditions ((D) through (F) and (J) through (L)). Iba-1 immunohistochemistry is shown at low and high magnification (left and middle columns, respectively). (M) through (O) The effect of RU486 (mifepristone (11β-[p-(dimethylamino)phenyl]-17β-hydroxy-17-(1-propynyl)estra-4,9-dien-3-one)) on the microglia population in response to LPS injection in stressed animals. Note that stress highly increased the microglial activation response to LPS injection (J) through (L) compared with nonstressed conditions (G) through (I). Note that, after LPS injection, most microglial cells display a round morphology typical of macrophages, whose density significantly increases under conditions of chronic stress. Also note how RU486 treatment strongly prevents the stress-induced sensitisation of microglia to subsequent LPS injection (M) through (O). The blue staining in all panels is the Monastral Blue inert tracer contained in the vehicle. Scale bars: 500 μm (A, D, G, J and M); 100 μm (all other panels). Abbreviations: V, Vehicle; S, Stress; SL, Lipopolysaccharide injected into stressed animals; SLR, Lipopolysaccharide injected into stressed animals treated with RU486.
Figure 4
Figure 4
Effect of lipopolysaccharide and stress on the number of activated microglial cells in the substantia nigra. Quantification of changes on the reactive microglial population in the substantia nigra at the end of the treatments. Results are mean ± SD of four independent experiments expressed as OX-6-positive cells/mm2. P < 0.001 by one-way analysis of variance followed by the least significant difference post hoc test for multiple range comparisons: a, compared with vehicle (V); b, compared with stress (S); c, compared with lipopolysaccharide (L); d, compared with stress + lipopolysaccharide (SL). SLR, Lipopolysaccharide injected into stressed animals treated with RU486 (mifepristone (11β-[p-(dimethylamino)phenyl]-17β-hydroxy-17-(1-propynyl)estra-4,9-dien-3-one)).
Figure 5
Figure 5
Effect of lipopolysaccharide and stress on expression of tumour necrosis factor α, interleukin 1β, interleukin 6 and inducible nitric oxide synthase mRNAs in substantia nigra. mRNA expression was quantified by real-time RT-PCR. Stress had no effect in the vehicle-injected animals. As expected, lipopolysaccharide (LPS) injection increased the expression levels of mRNA. This induction was higher when LPS and stress were combined, whereas treatment with RU486 (mifepristone (11β-[p-(dimethylamino)phenyl]-17β-hydroxy-17-(1-propynyl)estra-4,9-dien-3-one)) prevented this effect. Results are mean ± SD of at least three independent experiments expressed as percentage of control values. Statistical significance was calculated by one-way analysis of variance followed by the least significant difference post hoc test for multiple range comparisons a, compared with vehicle (V); b, compared with stress (S); c, compared with lipopolysaccharide (L); d, compared with stress + lipopolysaccharide (SL). SLR, Lipopolysaccharide injected into stressed animals treated with RU486 (mifepristone (11β-[p-(dimethylamino)phenyl]-17β-hydroxy-17-(1-propynyl)estra-4,9-dien-3-one)). (A) Tumour necrosis factor α (TNF-α), P < 0.001. (B) Interleukin 1β (IL-1β), P < 0.001. (C) Interleukin 6 (IL-6), P < 0.01. (D) Inducible nitric oxide synthase (iNOS), P < 0.01.
Figure 6
Figure 6
Effect of stress and lipopolysaccharide on inhibitor of nuclear factor κB kinase, subunit β, and Iba-1 in the substantia nigra. Iba-1 immunofluorescence (A), (D) and (G) and inhibitor of nuclear factor κB kinase, subunit β (IKKβ), immunofluorescence (B), (E) and (H) show robust induction of IKKβ in Iba-1-labelled microglial cells in the merged images (C), (F) and (I). Images (A), (B) and (C) were taken of an animal injected with lipopolysaccharide (LPS; L). Images (D) through (F) were taken of a stressed animal injected with LPS (SL). Images (G), (H) and (I) are high-magnification photomicrographs showing one representative cell. Scale bars: (A) through (F), 200 μm; (G) through (I), 25 μm. Arrows indicate colocalizing cells.
Figure 7
Figure 7
Effect of stress and lipopolysaccharide on lipid peroxidation in the substantia nigra. Lipid peroxidation increased in stressed animals as well as after the injection of lipopolysaccharide (LPS) in nonstressed animals. When combined, stress and LPS had an additive effect. P < 0.01 by one-way analysis of variance followed by least significant difference post hoc test for multiple range comparisons. a, compared with vehicle (V); b, compared with stress (S); c, compared with lipopolysaccharide (L). SL, stressed animals injected with lipopolysaccharide.
Figure 8
Figure 8
Effect of stress and lipopolysaccharide on the expression of CD200, chemokine (C-X3-C motif) receptor 1 and monocyte chemoattractant protein 1 mRNAs in the substantia nigra. mRNA expression was quantified by real-time RT-PCR. (A) CD200. Lipopolysaccharide (LPS) decreased CD200 expression in nonstressed animals and increased it in stressed rats. (B) Chemokine (C-X3-C motif) receptor 1 (CX3CR1). LPS decreased CX3CR1expression in nonstressed animals, and in stressed animals the decrease was even greater. (C) Monocyte chemoattractant protein 1 (MCP-1). No significant change was observed after injection of LPS into nonstressed rats. However, the effect of LPS on MCP-1 expression in stressed animals was massive. P < 0.01 by one-way analysis of variance followed by least significant difference post hoc test for multiple range comparisons. a, compared with vehicle (V); b, compared with lipopolysaccharide (L). SL, stressed animals injected with lipopolysaccharide.
Figure 9
Figure 9
Effect of stress and lipopolysaccharide in astroglia in the substantia nigra. (A) Coronal section showing glial fibrillary acidic protein (GFAP) immunoreactivity in a vehicle-injected nonstressed animal (arrow points to injection site). A limited alteration restricted to the needle tract is observed. (B) High-magnification image of the area within the box in (A). (C) GFAP immunoreactivity in a lipopolysaccharide (LPS)-injected nonstressed animal. There is an area lacking GFAP immunoreactivity around the injection track (dotted encircled area). (D) High-magnification image of the square box in (C); the arrow shows the injection site. (E) GFAP immunoreactivity in a LPS-injected stressed animal. The area lacking GFAP immunoreactivity is bigger (dotted encircled area). (F) High magnification of the square box in panel E showing hypertrophic astrocytes surrounding the injection site. Scale bars: (A), (C) and (E), 500 μm; (B), (D) and (F), 100 μm. Abbreviations: V, vehicle; S, stress; L, lipopolysaccharide; SL, lipopolysaccharide injected into stressed animals; SLR, lipopolysaccharide injected into stressed animals treated with RU486 (mifepristone (11β-[p-(dimethylamino)phenyl]-17β-hydroxy-17-(1-propynyl)estra-4,9-dien-3-one)). (G) Quantification of the areas lacking GFAP immunoreactivity on the substantia nigra at the end of the treatments. Results are mean ± SD of at least four independent experiments expressed in millimetres squared. P < 0.001 by analysis of variance followed by least significant difference post hoc test for multiple comparisons. a, compared with vehicle (V); b, compared with stress (S); c, compared with lipopolysaccharide (L). SL, stressed animals injected with lipopolysaccharide; SLR, lipopolysaccharide injected into stressed animals treated with RU486.
Figure 10
Figure 10
Chronic stress increases the lipopolysaccharide-induced loss of dopaminergic neurons in the substantia nigra. (A) Coronal section showing tyrosine hydroxylase (TH) immunoreactivity after the injection of vehicle (arrow) in nonstressed animals. (B) TH immunoreactivity after the injection of vehicle (arrow) in stressed animals. No significant changes can be observed. (C) TH immunoreactivity after the injection of 2 μg of lipopolysaccharide (LPS) into the substantia nigra (SN) of nonstressed rats. There is a loss of dopaminergic neurons around the injection track (arrow). (D) TH immunoreactivity after the injection of 2 μg of LPS into the SN of stressed rats. The loss of neurons is higher around the injection track (arrow). (E) RU486 (mifepristone (11β-[p-(dimethylamino)phenyl]-17β-hydroxy-17-(1-propynyl)estra-4,9-dien-3-one)) diminished the loss of TH-positive neurons caused by the combined action of LPS and stress. Scale bar: 500 μm. Abbreviations: V, vehicle; S, stress; L, lipopolysaccharide; SL, lipopolysaccharide injected into stressed animals; SLR, lipopolysaccharide injected into stressed animals treated with RU486. (F) Quantification of the number of TH-positive cells. Results are mean ± SD of four independent experiments expressed as TH-positive cells within the bounded area of the SN. P < 0.001 by analysis of variance followed by least significant difference post hoc test for multiple comparisons. a, compared with V; b, compared with S; c, compared with L; d, compared with SL. (G) Immunofluorescence of TH after the injection of 2 μg of LPS into the SN of stressed rats. (H) Immunofluorescence of Iba-1 after the injection of 2 μg of LPS into the SN of stressed rats. (I) Merged image of (G) and (H) showing activated microglia around the dopaminergic neurons. Scale bars in (G) through (I): 100 μm.

References

    1. Olanow CW, Shapira AHV, Agid Y. Neuroprotection for Parkinson’s disease: prospects and promises. Ann Neurol. 2003;53(Suppl 3):S1–S2.
    1. Dauer W, Przedborski S. Parkinson’s disease: mechanisms and models. Neuron. 2003;39:889–909. doi: 10.1016/S0896-6273(03)00568-3.
    1. Di Monte DA, Langston JW. In: Neuroglia. Kettenmann H, Ransom BR, editor. New York: Oxford University Press; 1995. Idiopathic and 1-methyl-4phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced Parkinsonism; pp. 989–997.
    1. Lesage S, Brice A. Parkinson’s disease: from monogenic forms to genetic susceptibility factors. Hum Mol Genet. 2009;18(R1):R48–R59. doi: 10.1093/hmg/ddp012.
    1. Gao HM, Liu B, Zhang W, Hong JS. Synergistic dopaminergic neurotoxicity of MPTP and inflammogen lipopolysaccharide: relevance to the etiology of Parkinson’s disease. FASEB J. 2003;17:1957–1959.
    1. Hirsch EC, Hunot S. Neuroinflammation in Parkinson’s disease: a target for neuroprotection? Lancet Neurol. 2009;8:382–397. doi: 10.1016/S1474-4422(09)70062-6.
    1. Jenner P. Oxidative stress in Parkinson’s disease. Ann Neurol. 2003;53(Suppl 3):S26–S38.
    1. Marchetti B, Abbracchio MP. To be or not to be (inflamed): Is that the question in anti-inflammatory drug therapy of neurodegenerative disorders? Trends Pharmacol Sci. 2005;26:517–525. doi: 10.1016/j.tips.2005.08.007.
    1. Marchetti B, Serra PA, L’Episcopo F, Tirolo C, Caniglia S, Testa N, Cioni S, Gennuso F, Rocchitta G, Desole MS, Mazzarino MC, Miele E, Morale MC. Hormones are key actors in gene × environment interactions programming vulnerability to Parkinson’s disease: glia as a common final pathway. Ann N Y Acad Sci. 2005;1057:296–318. doi: 10.1196/annals.1356.023.
    1. Marchetti B, Serra PA, Tirolo C, ’L’Episcopo F, Caniglia S, Gennuso F, Testa N, Miele E, Desole S, Barden N, Morale MC. Glucocorticoid receptor–nitric oxide crosstalk and vulnerability to experimental Parkinsonism: pivotal role for glia–neuron interactions. Brain Res Rev. 2005;48:302–321. doi: 10.1016/j.brainresrev.2004.12.030.
    1. McGeer PL, McGeer EG. Inflammation and neurodegeneration in Parkinson’s disease. Parkinsonism Relat Disord. 2004;10(Suppl 1):S3–S7.
    1. McGeer PL, Itagaki S, Boyes BE, McGeer EG. Reactive microglia are positive for HLA-DR in the substantia nigra of Parkinson’s and Alzheimer’s disease brains. Neurology. 1988;38:1285–1291. doi: 10.1212/WNL.38.8.1285.
    1. Klegeris A, McGeer EG, McGeer PL. Therapeutic approaches to inflammation in neurodegenerative disease. Curr Opin Neurol. 2007;20:351–357. doi: 10.1097/WCO.0b013e3280adc943.
    1. McGeer PL, McGeer EG. Glial reactions in Parkinson’s disease. Mov Disord. 2008;23:474–483. doi: 10.1002/mds.21751.
    1. Tansey MG, McCoy MK, Frank-Cannon TC. Neuroinflammatory mechanisms in Parkinson’s disease: potential environmental triggers, pathways, and targets for early therapeutic intervention. Exp Neurol. 2007;208:1–25. doi: 10.1016/j.expneurol.2007.07.004.
    1. Cicchetti F, Brownell AL, Williams K, Chen YI, Livni E, Isacson O. Neuroinflammation of the nigrostriatal pathway during progressive 6-OHDA dopamine degeneration in rats monitored by immunohistochemistry and PET imaging. Eur J Neurosci. 2002;15:991–998. doi: 10.1046/j.1460-9568.2002.01938.x.
    1. Gao HM, Jiang J, Wilson B, Zhang W, Hong JS, Liu B. Microglial activation-mediated delayed and progressive degeneration of rat nigral dopaminergic neurons: relevance to Parkinson’s disease. J Neurochem. 2002;81:1285–1297. doi: 10.1046/j.1471-4159.2002.00928.x.
    1. Liberatore GT, Jackson-Lewis V, Vukosavic S, Mandir AS, Vila M, McAuliffe WG, Dawson VL, Dawson TM, Przedborski S. Inducible nitric oxide synthase stimulates dopaminergic neurodegeneration in the MPTP model of Parkinson disease. Nat Med. 1999;5:1403–1409. doi: 10.1038/70978.
    1. Miklossy J, Doudet DD, Schwab C, Yu S, McGeer EG, McGeer PL. Role of ICAM-1 in persisting inflammation in Parkinson disease and MPTP monkeys. Exp Neurol. 2006;197:275–283. doi: 10.1016/j.expneurol.2005.10.034.
    1. Holst O, Ulmer AJ, Brade H, Flad HD, Rietschel ET. Biochemistry and cell biology of bacterial endotoxins. FEMS Immunol Med Microbiol. 1996;16:83–104. doi: 10.1111/j.1574-695X.1996.tb00126.x.
    1. Kielian TL, Blecha F. CD14 and other recognition molecules for lipopolysaccharide: a review. Immunopharmacology. 1995;29:187–205. doi: 10.1016/0162-3109(95)00003-C.
    1. Godoy M, Tarelli R, Ferrari CC, Sarchi MI, Pitossi FJ. Central and systemic IL-1 exacerbates neurodegeneration and motor symptoms in a model of Parkinson’s disease. Brain. 2008;131:1880–1894. doi: 10.1093/brain/awn101.
    1. Chen H, Jacobs E, Schwarzschild MA, McCullough ML, Calle EE, Thun MJ, Ascherio A. Nonsteroidal antiinflammatory drug use and the risk for Parkinson’s disease. Ann Neurol. 2005;58:963–967. doi: 10.1002/ana.20682.
    1. Chen H, Zhang S, Hernán MA, Schwarzschild MA, Willett WC, Colditz GA, Speizer FE, Ascherio A. Nonsteroidal anti-inflammatory drugs and the risk of Parkinson disease. Arch Neurol. 2003;60:1059–1064. doi: 10.1001/archneur.60.8.1059.
    1. Esposito E, Di Matteo V, Benigno A, Pierucci M, Crescimanno G, Di Giovanni G. Non-steroidal anti-inflammatory drugs in Parkinson’s disease. Exp Neurol. 2007;205:295–312. doi: 10.1016/j.expneurol.2007.02.008.
    1. Castaño A, Herrera AJ, Cano J, Machado A. Lipopolysaccharide intranigral injection induces inflammatory reaction and damage in nigrostriatal dopaminergic system. J Neurochem. 1998;70:1584–1592.
    1. Castaño A, Herrera AJ, Cano J, Machado A. The degenerative effect of a single intranigral injection of LPS on the dopaminergic system is prevented by dexamethasone, and not mimicked by rh-TNF-α, IL-1β and IFN-γ. J Neurochem. 2002;81:150–157. doi: 10.1046/j.1471-4159.2002.00799.x.
    1. Herrera AJ, Castaño A, Venero JL, Cano J, Machado A. The single intranigral injection of LPS as a new model for studying the selective effects of inflammatory reactions on dopaminergic system. Neurobiol Dis. 2000;7:429–447. doi: 10.1006/nbdi.2000.0289.
    1. Kim WG, Mohney RP, Wilson B, Jeohn GH, Liu B, Hong JS. Regional difference in susceptibility to lipopolysaccharide-induced neurotoxicity in the rat brain: role of microglia. J Neurosci. 2000;20:6309–6316.
    1. Gayle DA, Ling Z, Tong C, Landers T, Lipton JW, Carvey PM. Lipopolysaccharide (LPS)-induced dopamine cell loss in culture: roles of tumor necrosis factor-α, interleukin-1β, and nitric oxide. Brain Res Dev Brain Res. 2002;133:27–35. doi: 10.1016/S0165-3806(01)00315-7.
    1. Jeohn GH, Cooper CL, Wilson B, Chang RC, Jang KJ, Kim HC, Liu B, Hong JS. p38 MAP kinase is involved in lipopolysaccharide-induced dopaminergic neuronal cell death in rat mesencephalic neuron-glia cultures. Ann N Y Acad Sci. 2002;962:332–346. doi: 10.1111/j.1749-6632.2002.tb04078.x.
    1. Liu B, Du L, Hong JS. Naloxone protects rat dopaminergic neurons against inflammatory damage through inhibition of microglia activation and superoxide generation. J Pharmacol Exp Ther. 2000;293:607–617.
    1. Liu Y, Qin L, Li G, Zhang W, An L, Liu B, Hong JS. Dextromethorphan protects dopaminergic neurons against inflammation-mediated degeneration through inhibition of microglial activation. J Pharmacol Exp Ther. 2003;305:212–218. doi: 10.1124/jpet.102.043166.
    1. Foley P, Gerlach M, Double KL, Riederer P. Dopamine receptor agonists in the therapy of Parkinson’s disease. J Neural Transm. 2004;111:1375–1446. doi: 10.1007/s00702-003-0059-x.
    1. Caspi A, Sugden K, Moffitt TE, Taylor A, Craig IW, Harrington H, McClay J, Mill J, Martin J, Braithwaite A, Poulton R. Influence of life stress on depression: moderation by a polymorphism in the 5-HTT gene. Science. 2003;301:386–389. doi: 10.1126/science.1083968.
    1. Kendler KS, Karkowski-Shuman L. Stressful life events and genetic liability to major depression: genetic control of exposure to the environment? Psychol Med. 1997;27:539–547. doi: 10.1017/S0033291797004716.
    1. Charcot JM. Lectures on the Diseases of the Nervous System, Volume 1. London: The New Sydenham Society; 1878. (English translation by Sigerson G)
    1. Gowers WR. Diseases of the Nervous System. Philadelphia, PA: Blakiston, Son & Co; 1888. ((American edition)).
    1. McEwen BS. Protective and damaging effects of stress mediators: the good and bad sides of the response to stress. Metabolism. 2002;51(6 Suppl 1):2–4.
    1. McEwen BS. The neurobiology of stress: from serendipity to clinical relevance. Brain Res. 2000;886:172–189. doi: 10.1016/S0006-8993(00)02950-4.
    1. de Kloet ER, Joles M, Holsboer F. Stress and the brain: from adaptation to disease. Nat Rev Neurosci. 2005;6:463–475. doi: 10.1038/nrn1683.
    1. Joëls M, Krugers H, Karst H. Stress-induced changes in hippocampal function. Prog Brain Res. 2008;167:3–15.
    1. Espinosa-Oliva AM, de Pablos RM, Villarán RF, Arguelles S, Venero JL, Machado A, Cano J. Stress is critical for LPS-induced activation of microglia and damage in the rat hippocampus. Neurobiol Aging. 2011;32:85–102. doi: 10.1016/j.neurobiolaging.2009.01.012.
    1. de Pablos RM, Villaran RF, Argüelles S, Herrera AJ, Venero JL, Ayala A, Cano J, Machado A. Stress increases vulnerability to inflammation in the rat prefrontal cortex. J Neurosci. 2006;26:5709–5719. doi: 10.1523/JNEUROSCI.0802-06.2006.
    1. Paxinos G, Watson C. The Rat Brain in Stereotaxic Coordinates. London: Academic Press; 1986.
    1. Gamaro GD, Manoli P, Torres IL, Silveira R, Dalmaz C. Effects of chronic variate stress on feeding behavior and on monoamine levels in different rat brain structures. Neurochem Int. 2003;42:107–114. doi: 10.1016/S0197-0186(02)00080-3.
    1. Konarska M, Stewart RE, McCarty R. Predictability of chronic intermittent stress: effects on sympathetic-adrenal medullary responses of laboratory rats. Behav Neural Biol. 1990;53:231–243. doi: 10.1016/0163-1047(90)90458-I.
    1. Murua VS, Molina VA. Effects of chronic variable stress and antidepressant drugs on behavioral inactivity during an uncontrollable stress: interaction between both treatments. Behav Neural Biol. 1992;57:87–89. doi: 10.1016/0163-1047(92)90790-B.
    1. Muscat R, Papp M, Willner P. Reversal of stress-induced anhedonia by the atypical antidepressants, fluoxetine and maprotiline. Psychopharmacology (Berl) 1992;109:433–438. doi: 10.1007/BF02247719.
    1. Papp M, Willner P, Muscat R. An animal model of anhedonia: attenuation of sucrose consumption and place preference conditioning by chronic unpredictable mild stress. Psychopharmacology (Berl) 1991;104:255–259. doi: 10.1007/BF02244188.
    1. Willner P, Towell A, Sampson D, Sophokleous S, Muscat R. Reduction of sucrose preference by chronic unpredictable mild stress, and its restoration by a tricyclic antidepressant. Psychopharmacology (Berl) 1987;93:358–364.
    1. Gundersen HGJ, Bagger P, Bendtsen TF, Evans SM, Korbo L, Marcussen N, Møller A, Nielsen K, Nyengaard JR, Pakkenberg B, Sørensen FB, Vesterby A, West MJ. The new stereological tools: disector, fractionator, nucleator and point sampled intercepts and their use in pathological research and diagnosis. APMIS. 1988;96:857–881. doi: 10.1111/j.1699-0463.1988.tb00954.x.
    1. Meltzer JC, Sanders V, Grimm PC, Stern E, Rivier C, Lee S, Rennie SL, Gietz RD, Hole AK, Watson PH, Greenberg AH, Nance DM. Production of digoxigenin-labelled RNA probes and the detection of cytokine mRNA in rat spleen and brain by in situ hybridization. Brain Res Brain Res Protoc. 1998;2:339–351. doi: 10.1016/S1385-299X(98)00010-5.
    1. Lee DY, Oh YJ, Jin BK. Thrombin-activated microglia contributes to death of dopaminergic neurons in rat mesencephalic cultures: dual roles of mitogen-activated protein kinase signaling pathways. Glia. 2005;51:98–110. doi: 10.1002/glia.20190.
    1. Rostworowski M, Balasingam V, Chabot S, Owens T, Yong VW. Astrogliosis in the neonatal and adult murine brain post-trauma: elevation of inflammatory cytokines and the lack of requirement for endogenous interferon-γ. J Neurosci. 1997;17:3664–3674.
    1. Kim YW, Zhao RJ, Park SJ, Lee JR, Cho IJ, Yang CH, Kim SG, Kim SC. Anti-inflammatory effects of liquiritigenin as a consequence of the inhibition of NF-κB-dependent iNOS and pro-inflammatory cytokines production. Br J Pharmacol. 2008;154:165–173. doi: 10.1038/bjp.2008.79.
    1. Bland ST, Beckley JT, Watkins LR, Maier SF, Bilbo SD. Neonatal Escherichia coli infection alters glial, cytokine, and neuronal gene expression in response to acute amphetamine in adolescent rats. Neurosci Lett. 2010;474:52–57. doi: 10.1016/j.neulet.2010.03.006.
    1. Zhu J, Zhou Z, Liu Y, Zheng J. Fractalkine and CX3CR1 are involved in the migration of intravenously grafted human bone marrow stromal cells toward ischemic brain lesion in rats. Brain Res. 2009;1287:173–183.
    1. Fryer HJ, Davis GE, Manthorpe M, Varon S. Lowry protein assay using an automatic microtiter plate spectrophotometer. Anal Biochem. 1986;153:262–266. doi: 10.1016/0003-2697(86)90090-4.
    1. Jiang ZY, Woollard AC, Wolff SP. Lipid hydroperoxide measurement by oxidation of Fe2+ in the presence of xylenol orange: comparison with the TBA assay and an iodometric method. Lipids. 1991;26:853–856. doi: 10.1007/BF02536169.
    1. Tomás-Camardiel M, Venero JL, Herrera AJ, de Pablos RM, Pintor-Toro JA, Machado A, Cano J. Blood–brain barrier disruption highly induces aquaporin-4 mRNA and protein in perivascular and parenchymal astrocytes: protective effect by estradiol treatment in ovariectomized animals. J Neurosci Res. 2005;80:235–246. doi: 10.1002/jnr.20443.
    1. Deshmane SL, Kremlev S, Amini S, Sawaya BE. Monocyte chemoattractant protein-1 (MCP-1): an overview. J Interferon Cytokine Res. 2009;29:313–326. doi: 10.1089/jir.2008.0027.
    1. Hickman SE, El Khoury J. Mechanisms of mononuclear phagocyte recruitment in Alzheimer’s disease. CNS Neurol Disord Drug Targets. 2010;9:168–173. doi: 10.2174/187152710791011982.
    1. Morimoto M, Morita N, Ozawa H, Yokoyama K, Kawata M. Distribution of glucocorticoid receptor immunoreactivity and mRNA in the rat brain: an immunohistochemical and in situ hybridization study. Neurosci Res. 1996;26:235–269. doi: 10.1016/S0168-0102(96)01105-4.
    1. Venero JL, Burguillos MA, Brundin P, Joseph B. The executioners sing a new song: killer caspases activate microglia. Cell Death Differ. 2011;18:1679–1691. doi: 10.1038/cdd.2011.107.
    1. Lawson LJ, Perry VH, Dri P, Gordon S. Heterogeneity in the distribution and morphology of microglia in the normal adult mouse brain. Neuroscience. 1990;39:151–170. doi: 10.1016/0306-4522(90)90229-W.
    1. Smith LK, Jadavji NM, Colwell KL, Katrina Perehudoff S, Metz GA. Stress accelerates neural degeneration and exaggerates motor symptoms in a rat model of Parkinson’s disease. Eur J Neurosci. 2008;27:2133–2146. doi: 10.1111/j.1460-9568.2008.06177.x.
    1. Burguillos MA, Deierborg T, Kavanagh E, Persson A, Hajji N, Garcia-Quintanilla A, Cano J, Brundin P, Englund E, Venero JL, Joseph B. Caspase signalling controls microglia activation and neurotoxicity. Nature. 2011;472:319–324. doi: 10.1038/nature09788.
    1. Frank MG, Miguel ZD, Watkins L, Maier SF. Prior exposure to glucocorticoids sensitizes the neuroinflammatory and peripheral inflammatory responses to E. coli lipopolysaccharide. Brain Behav Immun. 2010;24:19–30. doi: 10.1016/j.bbi.2009.07.008.
    1. Frank MG, Thompson BM, Watkins LR, Maier SF. Glucocorticoids mediate stress-induced priming of microglial pro-inflammatory responses. Brain Behav Immun. 2012;26:337–345. doi: 10.1016/j.bbi.2011.10.005.
    1. Frank MG, Watkins LR, Maier SF. Stress- and glucocorticoid-induced priming of neuroinflammatory responses: potential mechanisms of stress-induced vulnerability to drugs of abuse. Brain Behav Immun. 2011;25(Suppl 1):S21–S28.
    1. Johnson JD, O’Connor KA, Deak T, Stark M, Watkins LR, Maier SF. Prior stressor exposure sensitizes LPS-induced cytokine production. Brain Behav Immun. 2002;16:461–476. doi: 10.1006/brbi.2001.0638.
    1. Perry VH. The influence of systemic inflammation on inflammation in the brain: implications for chronic neurodegenerative disease. Brain Behav Immun. 2004;18:407–413. doi: 10.1016/j.bbi.2004.01.004.
    1. Smyth GP, Stapleton PP, Freeman TA, Concannon EM, Mestre JR, Duff M, Maddali S, Daly JM. Glucocorticoid pretreatment induces cytokine overexpression and nuclear factor-κB activation in macrophages. J Surg Res. 2004;116:253–261. doi: 10.1016/S0022-4804(03)00300-7.
    1. Sorrells SF, Sapolsky RM. An inflammatory review of glucocorticoid actions in the CNS. Brain Behav Immun. 2007;21:259–272. doi: 10.1016/j.bbi.2006.11.006.
    1. Kierdorf K, Prinz M. Factors regulating microglia activation. Front Cell Neurosci. 2013;7:44.
    1. Conductier G, Blondeau N, Guyon A, Nahon JL, Rovère C. The role of monocyte chemoattractant protein MCP1/CCL2 in neuroinflammatory diseases. J Neuroimmunol. 2010;224:93–100. doi: 10.1016/j.jneuroim.2010.05.010.
    1. Barclay AN, Wright GJ, Brooke G, Brown MH. CD200 and membrane protein interactions in the control of myeloid cells. Trends Immunol. 2002;23:285–290. doi: 10.1016/S1471-4906(02)02223-8.
    1. Hoek RM, Ruuls SR, Murphy CA, Wright GJ, Goddard R, Zurawski SM, Blom B, Homola ME, Streit WJ, Brown MH, Barclay AN, Sedgwick JD. Down-regulation of the macrophage lineage through interaction with OX2 (CD200) Science. 2000;290:1768–1771.
    1. Biber K, Neumann H, Inoue K, Boddeke HW. Neuronal ‘On’ and ‘Off’ signals control microglia. Trends Neurosci. 2007;30:596–602. doi: 10.1016/j.tins.2007.08.007.
    1. Cardona AE, Pioro EP, Sasse ME, Kostenko V, Cardona SM, Dijkstra IM, Huang D, Kidd G, Dombrowski S, Dutta R, Lee JC, Cook DN, Jung S, Lira SA, Littman DR, Ransohoff RM. Control of microglial neurotoxicity by the fractalkine receptor. Nat Neurosci. 2006;9:917–924. doi: 10.1038/nn1715.
    1. Sozzani S, Zhou D, Locati M, Rieppi M, Proost P, Magazin M, Vita N, van Damme J, Mantovani A. Receptors and transduction pathways for monocyte chemotactic protein-2 and monocyte chemotactic protein-3: Similarities and differences with MCP-1. J Immunol. 1994;152:3615–3622.
    1. Stamatovic SM, Keep RF, Kunkel SL, Andjelkovic AV. Potential role of MCP-1 in endothelial cell tight junction ‘opening’: signaling via Rho and Rho kinase. J Cell Sci. 2003;116:4615–4628. doi: 10.1242/jcs.00755.
    1. Ataka K, Asakawa A, Nagaishi K, Kaimoto K, Sawada A, Hayakawa Y, Tatezawa R, Inui A, Fujimiya M. Bone marrow-derived microglia infiltrate into the paraventricular nucleus of chronic psychological stress-loaded mice. PLoS One. 2013;8:e81744. doi: 10.1371/journal.pone.0081744.
    1. Calvez J, Fromentin G, Nadkarni N, Darcel N, Even P, Tomé D, Ballet N, Chaumontet C. Inhibition of food intake induced by acute stress in rats is due to satiation effects. Physiol Behav. 2011;104:675–683. doi: 10.1016/j.physbeh.2011.07.012.
    1. Nair A, Bonneau RH. Stress-induced elevation of glucocorticoids increases microglia proliferation through NMDA receptor activation. J Neuroimmunol. 2006;171:72–85. doi: 10.1016/j.jneuroim.2005.09.012.
    1. Ros-Bernal F, Hunot S, Herrero MT, Parnadeau S, Corvol JC, Lu L, Alvarez-Fischer D, Carrillo-de Sauvage MA, Saurini F, Coussieu C, Kinugawa K, Prigent A, Höglinger G, Hamon M, Tronche F, Hirsch EC, Vyas S. Microglial glucocorticoid receptors play a pivotal role in regulating dopaminergic neurodegeneration in parkinsonism. Proc Natl Acad Sci USA. 2011;108:6632–6637. doi: 10.1073/pnas.1017820108.
    1. Kim YM, Lee JY, Choi SH, Kim DG, Jahng JW. RU486 blocks fasting-induced decrease of neuronal nitric oxide synthase in the rat paraventricular nucleus. Brain Res. 2004;1018:221–226. doi: 10.1016/j.brainres.2004.05.068.
    1. Mailliet F, Qi H, Rocher C, Spedding M, Svenningsson P, Jay TM. Protection of stress-induced impairment of hippocampal/prefrontal LTP through blockade of glucocorticoid receptors: implication of MEK signaling. Exp Neurol. 2008;211:593–596. doi: 10.1016/j.expneurol.2008.02.030.
    1. Sharrett-Field L, Butler TR, Berry JN, Reynolds AR, Prendergast MA. Mifepristone pretreatment reduces ethanol withdrawal severity in vivo. Alcohol Clin Exp Res. 2013;37:1417–1423. doi: 10.1111/acer.12093.
    1. Ahima RS, Harlan RE. Charting of type II glucocorticoid receptor-like immunoreactivity in the rat central nervous system. Neuroscience. 1990;39:579–604. doi: 10.1016/0306-4522(90)90244-X.
    1. Ahima RS, Tagoe CNB, Harlan RE. Type II corticosteroid receptor-like immunoreactivity in the rat cerebellar cortex: differential regulation by corticosterone. Neuroendocrinology. 1992;55:683–694. doi: 10.1159/000126188.
    1. Maki BE, McIlroy WE. Influence of arousal and attention on the control of postural sway. J Vestib Res. 1996;6:53–59.
    1. Metz GA, Jadavji NM, Smith LK. Modulation of motor function by stress: a novel concept of the effects of stress on behavior. Eur J Neurosci. 2005;22:1190–1200. doi: 10.1111/j.1460-9568.2005.04285.x.
    1. Metz GAS, Schwab ME, Welzl H. The effects of acute and chronic stress on motor and sensory performance in male Lewis rats. Physiol Behav. 2001;72:29–35. doi: 10.1016/S0031-9384(00)00371-1.
    1. Charlett A, Dobbs RJ, Purkiss AG, Wright DJ, Peterson DW, Weller C, Dobbs SM. Cortisol is higher in parkinsonism and associated with gait deficit. Acta Neurol Scand. 1998;97:77–85.
    1. Kim ST, Choi JH, Chang JW, Kim SW, Hwang O. Immobilization stress causes increases in tetrahydrobiopterin, dopamine, and neuromelanin and oxidative damage in the nigrostriatal system. J Neurochem. 2005;95:89–98. doi: 10.1111/j.1471-4159.2005.03342.x.
    1. Finlay JM, Zigmond MJ. The effects of stress on central dopaminergic neurons: possible clinical implications. Neurochem Res. 1997;22:1387–1394. doi: 10.1023/A:1022075324164.
    1. Izzo E, Sanna PP, Koob GF. Impairment of dopaminergic system function after chronic treatment with corticotropin-releasing factor. Pharmacol Biochem Behav. 2005;81:701–708. doi: 10.1016/j.pbb.2005.04.017.
    1. Pani L, Porcella A, Gessa GL. The role of stress in the pathophysiology of the dopaminergic system. Mol Psychiatry. 2000;5:14–21. doi: 10.1038/sj.mp.4000589.
    1. Smith AD, Castro SL, Zigmond MJ. Stress-induced Parkinson’s disease: a working hypothesis. Physiol Behav. 2002;77:527–531. doi: 10.1016/S0031-9384(02)00939-3.
    1. Zigmond MJ, Abercrombie ED, Berger TW, Grace AA, Stricker EM. In: Current Concepts in Parkinson’s Disease Research. Schneider JS, Gupta M, editor. Toronto: Hogrefe & Huber; 1993. Compensatory responses to partial loss of dopaminergic neurons: studies with 6-hydroxydopamine; pp. 99–140.
    1. Stein-Behrens BA, Elliot EM, Miller CA, Schilling JW, Newcombe R, Sapolsky RM. Glucocorticoids exacerbate kainic acid-induced extracellular accumulation of excitatory amino acids in the rat hippocampus. J Neurochem. 1992;58:1730–1735. doi: 10.1111/j.1471-4159.1992.tb10047.x.

Source: PubMed

3
Abonneren