Skip to main content
Top
Published in: Journal of Neuroinflammation 1/2017

Open Access 01-12-2017 | Research

Lack of interleukin-13 receptor α1 delays the loss of dopaminergic neurons during chronic stress

Authors: Simone Mori, Shuei Sugama, William Nguyen, Tatiana Michel, M. Germana Sanna, Manuel Sanchez-Alavez, Rigo Cintron-Colon, Gianluca Moroncini, Yoshihiko Kakinuma, Pamela Maher, Bruno Conti

Published in: Journal of Neuroinflammation | Issue 1/2017

Login to get access

Abstract

Background

The majority of Parkinson’s disease (PD) cases are sporadic and idiopathic suggesting that this neurodegenerative disorder is the result of both environmental and genetic factors. Stress and neuroinflammation are among the factors being investigated for their possible contributions to PD. Experiments in rodents showed that severe chronic stress can reduce the number of dopaminergic neurons in the substantia nigra pars compacta (SNc); the same cells that are lost in PD. These actions are at least in part mediated by increased oxidative stress. Here, we tested the hypothesis that the interleukin-13 receptor alpha 1 (IL-13Rα1), a cytokine receptor whose activation increases the vulnerability of dopaminergic neurons to oxidative damage, participates in the stress-dependent damage of these neurons.

Methods

Mice were subject to daily sessions of 8 h (acute) stress for 16 weeks (5 days a week), a procedure previously showed to induce loss of dopaminergic neurons in the SNc. The source and the kinetics of interleukin-13 (IL-13), the endogenous ligand of IL-13Rα1, were evaluated 0, 1, 3, 6, and 8 h and at 16 weeks of stress. Identification of IL-13 producing cell-type was performed by immunofluorescent and by in situ hybridization experiments. Markers of oxidative stress, microglia activation, and the number of dopaminergic neurons in IL-13Rα1 knock-out animals (Il13ra1 Y/ ) and their wild-type littermates (Il13ra1 Y/+ ) were evaluated at 16 weeks of stress and at 20 weeks, following a 4 week non-stressed period and compared to non-stressed mice.

Results

IL-13 was expressed in microglial cells within the SN and in a fraction of the tyrosine hydroxylase-positive neurons in the SNc. IL-13 levels were elevated during daily stress and peaked at 6 h. 16 weeks of chronic restraint stress significantly reduced the number of SNc dopaminergic neurons in Il13ra1 Y/+ mice. Neuronal loss at 16 weeks was significantly lower in Il13ra1 Y/− mice. However, the loss of dopaminergic neurons measured at 20 weeks, after 4 weeks of non-stress following the 16 weeks of stress, was similar in Il13ra1 Y/+ and Il13ra1 Y/− mice.

Conclusions

IL-13, a cytokine previously demonstrated to increase the susceptibility of SNc dopaminergic neurons to oxidative stress, is elevated in the SN by restraint stress. Lack of IL-13Rα1 did not prevent nor halted but delayed neuronal loss in the mouse model of chronic restraint stress. IL-13/IL-13Rα1 may represent a target to reduce the rate of DA neuronal loss that can occur during severe chronic restraint stress.
Appendix
Available only for authorised users
Literature
1.
3.
go back to reference Sapolsky RM. Stress, glucocorticoids, and damage to the nervous system: the current state of confusion. Stress. 1996;1:1–19.CrossRefPubMed Sapolsky RM. Stress, glucocorticoids, and damage to the nervous system: the current state of confusion. Stress. 1996;1:1–19.CrossRefPubMed
4.
go back to reference Sapolsky RM, Uno H, Rebert CS, Finch CE. Hippocampal damage associated with prolonged glucocorticoid exposure in primates. J Neurosci. 1990;10:2897–902.PubMed Sapolsky RM, Uno H, Rebert CS, Finch CE. Hippocampal damage associated with prolonged glucocorticoid exposure in primates. J Neurosci. 1990;10:2897–902.PubMed
5.
go back to reference Sanchez-Alavez M, Conti B, Moroncini G, Criado JR. Contributions of neuronal prion protein on sleep recovery and stress response following sleep deprivation. Brain Res. 2007;1158:71–80.CrossRefPubMedPubMedCentral Sanchez-Alavez M, Conti B, Moroncini G, Criado JR. Contributions of neuronal prion protein on sleep recovery and stress response following sleep deprivation. Brain Res. 2007;1158:71–80.CrossRefPubMedPubMedCentral
6.
go back to reference Gibberd FB, Simmonds JP. Neurological disease in ex-far-east prisoners of war. Lancet. 1980;2:135–7.CrossRefPubMed Gibberd FB, Simmonds JP. Neurological disease in ex-far-east prisoners of war. Lancet. 1980;2:135–7.CrossRefPubMed
7.
go back to reference Hemmerle AM, Herman JP, Seroogy KB. Stress, depression and Parkinson’s disease. Exp Neurol. 2012;233:79–86.CrossRefPubMed Hemmerle AM, Herman JP, Seroogy KB. Stress, depression and Parkinson’s disease. Exp Neurol. 2012;233:79–86.CrossRefPubMed
8.
go back to reference Smith AD, Castro SL, Zigmond MJ. Stress-induced Parkinson’s disease: a working hypothesis. Physiol Behav. 2002;77:527–31.CrossRefPubMed Smith AD, Castro SL, Zigmond MJ. Stress-induced Parkinson’s disease: a working hypothesis. Physiol Behav. 2002;77:527–31.CrossRefPubMed
9.
go back to reference 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;27:2133–46.CrossRefPubMedPubMedCentral 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;27:2133–46.CrossRefPubMedPubMedCentral
10.
go back to reference Sugama S, Kakinuma Y. Loss of dopaminergic neurons occurs in the ventral tegmental area and hypothalamus of rats following chronic stress: possible pathogenetic loci for depression involved in Parkinson’s disease. Neurosci Res. 2016;111:48–55.CrossRefPubMed Sugama S, Kakinuma Y. Loss of dopaminergic neurons occurs in the ventral tegmental area and hypothalamus of rats following chronic stress: possible pathogenetic loci for depression involved in Parkinson’s disease. Neurosci Res. 2016;111:48–55.CrossRefPubMed
11.
go back to reference Sugama S, Sekiyama K, Kodama T, Takamatsu Y, Takenouchi T, Hashimoto M, Bruno C, Kakinuma Y. Chronic restraint stress triggers dopaminergic and noradrenergic neurodegeneration: possible role of chronic stress in the onset of Parkinson’s disease. Brain Behav Immun. 2016;51:39–46.CrossRefPubMed Sugama S, Sekiyama K, Kodama T, Takamatsu Y, Takenouchi T, Hashimoto M, Bruno C, Kakinuma Y. Chronic restraint stress triggers dopaminergic and noradrenergic neurodegeneration: possible role of chronic stress in the onset of Parkinson’s disease. Brain Behav Immun. 2016;51:39–46.CrossRefPubMed
12.
go back to reference Hemmerle AM, Dickerson JW, Herman JP, Seroogy KB. Stress exacerbates experimental Parkinson’s disease. Mol Psychiatry. 2014;19:638–40.CrossRefPubMed Hemmerle AM, Dickerson JW, Herman JP, Seroogy KB. Stress exacerbates experimental Parkinson’s disease. Mol Psychiatry. 2014;19:638–40.CrossRefPubMed
13.
go back to reference de Pablos RM, Herrera AJ, Espinosa-Oliva AM, Sarmiento M, Munoz MF, Machado A, Venero JL. Chronic stress enhances microglia activation and exacerbates death of nigral dopaminergic neurons under conditions of inflammation. J Neuroinflammation. 2014;11:34.CrossRefPubMedPubMedCentral de Pablos RM, Herrera AJ, Espinosa-Oliva AM, Sarmiento M, Munoz MF, Machado A, Venero JL. Chronic stress enhances microglia activation and exacerbates death of nigral dopaminergic neurons under conditions of inflammation. J Neuroinflammation. 2014;11:34.CrossRefPubMedPubMedCentral
14.
go back to reference Ong LK, Zhao Z, Kluge M, Tebay C, Zalewska K, Dickson PW, Johnson SJ, Nilsson M, Walker FR. Reconsidering the role of glial cells in chronic stress-induced dopaminergic neurons loss within the substantia nigra? Friend or foe? Brain Behav Immun. 2017;60:117–25.CrossRefPubMed Ong LK, Zhao Z, Kluge M, Tebay C, Zalewska K, Dickson PW, Johnson SJ, Nilsson M, Walker FR. Reconsidering the role of glial cells in chronic stress-induced dopaminergic neurons loss within the substantia nigra? Friend or foe? Brain Behav Immun. 2017;60:117–25.CrossRefPubMed
15.
go back to reference Sugama S, Fujita M, Hashimoto M, Conti B. Stress induced morphological microglial activation in the rodent brain: involvement of interleukin-18. Neuroscience. 2007;146:1388–99.CrossRefPubMed Sugama S, Fujita M, Hashimoto M, Conti B. Stress induced morphological microglial activation in the rodent brain: involvement of interleukin-18. Neuroscience. 2007;146:1388–99.CrossRefPubMed
16.
go back to reference Hinwood M, Morandini J, Day TA, Walker FR. Evidence that microglia mediate the neurobiological effects of chronic psychological stress on the medial prefrontal cortex. Cereb Cortex. 2012;22:1442–54.CrossRefPubMed Hinwood M, Morandini J, Day TA, Walker FR. Evidence that microglia mediate the neurobiological effects of chronic psychological stress on the medial prefrontal cortex. Cereb Cortex. 2012;22:1442–54.CrossRefPubMed
17.
go back to reference Tynan RJ, Naicker S, Hinwood M, Nalivaiko E, Buller KM, Pow DV, Day TA, Walker FR. Chronic stress alters the density and morphology of microglia in a subset of stress-responsive brain regions. Brain Behav Immun. 2010;24:1058–68.CrossRefPubMed Tynan RJ, Naicker S, Hinwood M, Nalivaiko E, Buller KM, Pow DV, Day TA, Walker FR. Chronic stress alters the density and morphology of microglia in a subset of stress-responsive brain regions. Brain Behav Immun. 2010;24:1058–68.CrossRefPubMed
18.
go back to reference Sugama S, Takenouchi T, Fujita M, Conti B, Hashimoto M. Differential microglial activation between acute stress and lipopolysaccharide treatment. J Neuroimmunol. 2009;207:24–31. Sugama S, Takenouchi T, Fujita M, Conti B, Hashimoto M. Differential microglial activation between acute stress and lipopolysaccharide treatment. J Neuroimmunol. 2009;207:24–31.
19.
go back to reference Diz-Chaves Y, Pernia O, Carrero P, Garcia-Segura LM. Prenatal stress causes alterations in the morphology of microglia and the inflammatory response of the hippocampus of adult female mice. J Neuroinflammation. 2012;9:71.CrossRefPubMedPubMedCentral Diz-Chaves Y, Pernia O, Carrero P, Garcia-Segura LM. Prenatal stress causes alterations in the morphology of microglia and the inflammatory response of the hippocampus of adult female mice. J Neuroinflammation. 2012;9:71.CrossRefPubMedPubMedCentral
20.
go back to reference Kojo A, Yamada K, Kubo KY, Yamashita A, Yamamoto T. Occlusal disharmony in mice transiently activates microglia in hippocampal CA1 region but not in dentate gyrus. Tohoku J Exp Med. 2010;221:237–43.CrossRefPubMed Kojo A, Yamada K, Kubo KY, Yamashita A, Yamamoto T. Occlusal disharmony in mice transiently activates microglia in hippocampal CA1 region but not in dentate gyrus. Tohoku J Exp Med. 2010;221:237–43.CrossRefPubMed
21.
go back to reference Sugama S, Takenouchi T, Fujita M, Kitani H, Hashimoto M. Cold stress induced morphological microglial activation and increased IL-1beta expression in astroglial cells in rat brain. J Neuroimmunol. 2011;233:29–36.CrossRefPubMed Sugama S, Takenouchi T, Fujita M, Kitani H, Hashimoto M. Cold stress induced morphological microglial activation and increased IL-1beta expression in astroglial cells in rat brain. J Neuroimmunol. 2011;233:29–36.CrossRefPubMed
22.
go back to reference Mcgeer PL, Itagaki S, Akiyama H, Mcgeer EG. Rate of cell death in Parkinsonism indicates active neuropathological process. Ann Neurol. 1988;24:574–6.CrossRefPubMed Mcgeer PL, Itagaki S, Akiyama H, Mcgeer EG. Rate of cell death in Parkinsonism indicates active neuropathological process. Ann Neurol. 1988;24:574–6.CrossRefPubMed
23.
go back to reference Perez Nievas BG, Hammerschmidt T, Kummer MP, Terwel D, Leza JC, Heneka MT. Restraint stress increases neuroinflammation independently of amyloid beta levels in amyloid precursor protein/PS1 transgenic mice. J Neurochem. 2011;116:43–52.CrossRefPubMed Perez Nievas BG, Hammerschmidt T, Kummer MP, Terwel D, Leza JC, Heneka MT. Restraint stress increases neuroinflammation independently of amyloid beta levels in amyloid precursor protein/PS1 transgenic mice. J Neurochem. 2011;116:43–52.CrossRefPubMed
25.
go back to reference Tansey MG, Goldberg MS. Neuroinflammation in Parkinson’s disease: its role in neuronal death and implications for therapeutic intervention. Neurobiol Dis. 2010;37:510–8.CrossRefPubMed Tansey MG, Goldberg MS. Neuroinflammation in Parkinson’s disease: its role in neuronal death and implications for therapeutic intervention. Neurobiol Dis. 2010;37:510–8.CrossRefPubMed
26.
go back to reference Morrison BE, Marcondes MC, Nomura DK, Sanchez-Alavez M, Sanchez-Gonzalez A, Saar I, Kim KS, Bartfai T, Maher P, Sugama S, Conti B. Cutting edge: IL-13ralpha1 expression in dopaminergic neurons contributes to their oxidative stress-mediated loss following chronic peripheral treatment with lipopolysaccharide. J Immunol. 2012;189:5498–502.CrossRefPubMedPubMedCentral Morrison BE, Marcondes MC, Nomura DK, Sanchez-Alavez M, Sanchez-Gonzalez A, Saar I, Kim KS, Bartfai T, Maher P, Sugama S, Conti B. Cutting edge: IL-13ralpha1 expression in dopaminergic neurons contributes to their oxidative stress-mediated loss following chronic peripheral treatment with lipopolysaccharide. J Immunol. 2012;189:5498–502.CrossRefPubMedPubMedCentral
28.
go back to reference Ramalingam TR, Pesce JT, Sheikh F, Cheever AW, Mentink-Kane MM, Wilson MS, Stevens S, Valenzuela DM, Murphy AJ, Yancopoulos GD, et al. Unique functions of the type II interleukin 4 receptor identified in mice lacking the interleukin 13 receptor alpha1 chain. Nat Immunol. 2008;9:25–33.CrossRefPubMed Ramalingam TR, Pesce JT, Sheikh F, Cheever AW, Mentink-Kane MM, Wilson MS, Stevens S, Valenzuela DM, Murphy AJ, Yancopoulos GD, et al. Unique functions of the type II interleukin 4 receptor identified in mice lacking the interleukin 13 receptor alpha1 chain. Nat Immunol. 2008;9:25–33.CrossRefPubMed
29.
go back to reference Volpe BT, Blau AD, Wessel TC, Saji M. Delayed histopathological neuronal damage in the substantia nigra compacta (nucleus A9) after transient forebrain ischaemia. Neurobiol Dis. 1995;2:119–27.CrossRefPubMed Volpe BT, Blau AD, Wessel TC, Saji M. Delayed histopathological neuronal damage in the substantia nigra compacta (nucleus A9) after transient forebrain ischaemia. Neurobiol Dis. 1995;2:119–27.CrossRefPubMed
30.
go back to reference Volpe BT, Wildmann J, Altar CA. Brain-derived neurotrophic factor prevents the loss of nigral neurons induced by excitotoxic striatal-pallidal lesions. Neuroscience. 1998;83:741–8.CrossRefPubMed Volpe BT, Wildmann J, Altar CA. Brain-derived neurotrophic factor prevents the loss of nigral neurons induced by excitotoxic striatal-pallidal lesions. Neuroscience. 1998;83:741–8.CrossRefPubMed
31.
go back to reference Yu JT, Lee CH, Yoo KY, Choi JH, Li H, Park OK, Yan B, Hwang IK, Kwon YG, Kim YM, Won MH. Maintenance of anti-inflammatory cytokines and reduction of glial activation in the ischemic hippocampal CA1 region preconditioned with lipopolysaccharide. J Neurol Sci. 2010;296:69–78.CrossRefPubMed Yu JT, Lee CH, Yoo KY, Choi JH, Li H, Park OK, Yan B, Hwang IK, Kwon YG, Kim YM, Won MH. Maintenance of anti-inflammatory cytokines and reduction of glial activation in the ischemic hippocampal CA1 region preconditioned with lipopolysaccharide. J Neurol Sci. 2010;296:69–78.CrossRefPubMed
32.
go back to reference Zhao X, Wang H, Sun G, Zhang J, Edwards NJ, Aronowski J. Neuronal interleukin-4 as a modulator of microglial pathways and ischemic brain damage. J Neurosci. 2015;35:11281–91.CrossRefPubMedPubMedCentral Zhao X, Wang H, Sun G, Zhang J, Edwards NJ, Aronowski J. Neuronal interleukin-4 as a modulator of microglial pathways and ischemic brain damage. J Neurosci. 2015;35:11281–91.CrossRefPubMedPubMedCentral
33.
go back to reference Brombacher TM, Nono JK, De Gouveia KS, Makena N, Darby M, Womersley J, Tamgue O, Brombacher F. IL-13-mediated regulation of learning and memory. J Immunol. 2017;198(7):2681–8.PubMed Brombacher TM, Nono JK, De Gouveia KS, Makena N, Darby M, Womersley J, Tamgue O, Brombacher F. IL-13-mediated regulation of learning and memory. J Immunol. 2017;198(7):2681–8.PubMed
34.
go back to reference Fontella FU, Cimarosti H, Crema LM, Thomazi AP, Leite MC, Salbego C, Goncalves CA, Wofchuk S, Dalmaz C, Netto CA. Acute and repeated restraint stress influences cellular damage in rat hippocampal slices exposed to oxygen and glucose deprivation. Brain Res Bull. 2005;65:443–50.CrossRefPubMed Fontella FU, Cimarosti H, Crema LM, Thomazi AP, Leite MC, Salbego C, Goncalves CA, Wofchuk S, Dalmaz C, Netto CA. Acute and repeated restraint stress influences cellular damage in rat hippocampal slices exposed to oxygen and glucose deprivation. Brain Res Bull. 2005;65:443–50.CrossRefPubMed
35.
go back to reference Fontella FU, Siqueira IR, Vasconcellos AP, Tabajara AS, Netto CA, Dalmaz C. Repeated restraint stress induces oxidative damage in rat hippocampus. Neurochem Res. 2005;30:105–11.CrossRefPubMed Fontella FU, Siqueira IR, Vasconcellos AP, Tabajara AS, Netto CA, Dalmaz C. Repeated restraint stress induces oxidative damage in rat hippocampus. Neurochem Res. 2005;30:105–11.CrossRefPubMed
36.
go back to reference Kreisel T, Frank MG, Licht T, Reshef R, Ben-Menachem-Zidon O, Baratta MV, Maier SF, Yirmiya R. Dynamic microglial alterations underlie stress-induced depressive-like behavior and suppressed neurogenesis. Mol Psychiatry. 2014;19:699–709.CrossRefPubMed Kreisel T, Frank MG, Licht T, Reshef R, Ben-Menachem-Zidon O, Baratta MV, Maier SF, Yirmiya R. Dynamic microglial alterations underlie stress-induced depressive-like behavior and suppressed neurogenesis. Mol Psychiatry. 2014;19:699–709.CrossRefPubMed
37.
go back to reference Bollinger JL, Bergeon Burns CM, Wellman CL. Differential effects of stress on microglial cell activation in male and female medial prefrontal cortex. Brain Behav Immun. 2016;52:88–97.CrossRefPubMed Bollinger JL, Bergeon Burns CM, Wellman CL. Differential effects of stress on microglial cell activation in male and female medial prefrontal cortex. Brain Behav Immun. 2016;52:88–97.CrossRefPubMed
38.
go back to reference Rasheed N, Ahmad A, Pandey CP, Chaturvedi RK, Lohani M, Palit G. Differential response of central dopaminergic system in acute and chronic unpredictable stress models in rats. Neurochem Res. 2010;35:22–32.CrossRefPubMed Rasheed N, Ahmad A, Pandey CP, Chaturvedi RK, Lohani M, Palit G. Differential response of central dopaminergic system in acute and chronic unpredictable stress models in rats. Neurochem Res. 2010;35:22–32.CrossRefPubMed
39.
go back to reference Mizoguchi K, Yuzurihara M, Ishige A, Sasaki H, Chui DH, Tabira T. Chronic stress induces impairment of spatial working memory because of prefrontal dopaminergic dysfunction. J Neurosci. 2000;20:1568–74.PubMed Mizoguchi K, Yuzurihara M, Ishige A, Sasaki H, Chui DH, Tabira T. Chronic stress induces impairment of spatial working memory because of prefrontal dopaminergic dysfunction. J Neurosci. 2000;20:1568–74.PubMed
40.
go back to reference Watanabe Y, Gould E, McEwen BS. Stress induces atrophy of apical dendrites of hippocampal CA3 pyramidal neurons. Brain Res. 1992;588:341–5.CrossRefPubMed Watanabe Y, Gould E, McEwen BS. Stress induces atrophy of apical dendrites of hippocampal CA3 pyramidal neurons. Brain Res. 1992;588:341–5.CrossRefPubMed
41.
go back to reference Monteiro S, Roque S, de Sa-Calcada D, Sousa N, Correia-Neves M, Cerqueira JJ. An efficient chronic unpredictable stress protocol to induce stress-related responses in C57BL/6 mice. Front Psych. 2015;6:6. Monteiro S, Roque S, de Sa-Calcada D, Sousa N, Correia-Neves M, Cerqueira JJ. An efficient chronic unpredictable stress protocol to induce stress-related responses in C57BL/6 mice. Front Psych. 2015;6:6.
42.
go back to reference Bardin L, Malfetes N, Newman-Tancredi A, Depoortere R. Chronic restraint stress induces mechanical and cold allodynia, and enhances inflammatory pain in rat: relevance to human stress-associated painful pathologies. Behav Brain Res. 2009;205:360–6.CrossRefPubMed Bardin L, Malfetes N, Newman-Tancredi A, Depoortere R. Chronic restraint stress induces mechanical and cold allodynia, and enhances inflammatory pain in rat: relevance to human stress-associated painful pathologies. Behav Brain Res. 2009;205:360–6.CrossRefPubMed
43.
go back to reference Voorhees JL, Tarr AJ, Wohleb ES, Godbout JP, Mo X, Sheridan JF, Eubank TD, Marsh CB. Prolonged restraint stress increases IL-6, reduces IL-10, and causes persistent depressive-like behavior that is reversed by recombinant IL-10. Plos One. 2013;8:e58488.CrossRefPubMedPubMedCentral Voorhees JL, Tarr AJ, Wohleb ES, Godbout JP, Mo X, Sheridan JF, Eubank TD, Marsh CB. Prolonged restraint stress increases IL-6, reduces IL-10, and causes persistent depressive-like behavior that is reversed by recombinant IL-10. Plos One. 2013;8:e58488.CrossRefPubMedPubMedCentral
44.
go back to reference Conti B, Sanchez-Alavez M, Winsky-Sommerer R, Morale MC, Lucero J, Brownell S, Fabre V, Huitron-Resendiz S, Henriksen S, Zorrilla EP, et al. Transgenic mice with a reduced core body temperature have an increased life span. Science. 2006;314:825–8.CrossRefPubMed Conti B, Sanchez-Alavez M, Winsky-Sommerer R, Morale MC, Lucero J, Brownell S, Fabre V, Huitron-Resendiz S, Henriksen S, Zorrilla EP, et al. Transgenic mice with a reduced core body temperature have an increased life span. Science. 2006;314:825–8.CrossRefPubMed
Metadata
Title
Lack of interleukin-13 receptor α1 delays the loss of dopaminergic neurons during chronic stress
Authors
Simone Mori
Shuei Sugama
William Nguyen
Tatiana Michel
M. Germana Sanna
Manuel Sanchez-Alavez
Rigo Cintron-Colon
Gianluca Moroncini
Yoshihiko Kakinuma
Pamela Maher
Bruno Conti
Publication date
01-12-2017
Publisher
BioMed Central
Published in
Journal of Neuroinflammation / Issue 1/2017
Electronic ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-017-0862-1

Other articles of this Issue 1/2017

Journal of Neuroinflammation 1/2017 Go to the issue