Skip to main content
Top
Published in: Journal of Neuroinflammation 1/2019

Open Access 01-12-2019 | Alzheimer's Disease | Research

Soluble epoxide hydrolase modulates immune responses in activated astrocytes involving regulation of STAT3 activity

Authors: Chia-Chi Hung, Yi-Hsuan Lee, Yi-Min Kuo, Pei-Chien Hsu, Huey-Jen Tsay, Ying-Ting Hsu, Chih-Chin Lee, Jia-Jun Liang, Feng-Shiun Shie

Published in: Journal of Neuroinflammation | Issue 1/2019

Login to get access

Abstract

Background

Astrocyte activation is a common pathological feature in many brain diseases with neuroinflammation, and revealing the underlying mechanisms might shed light on the regulatory processes of the diseases. Recently, soluble epoxide hydrolase (sEH) has been proposed to affect neuroinflammation in brain injuries. However, the roles of astrocytic sEH in brains with neurodegeneration remain unclear.

Methods

The expression of astrocytic sEH in the brains of APPswe/PSEN1dE9 (APP/PS1) mice developing Alzheimer’s disease (AD)-like pathology was evaluated by confocal imaging. LPS-activated primary astrocytes with mRNA silencing or overexpression of sEH were used to investigate its regulatory roles in astrocyte activation and the induction of pro-inflammatory markers. Primary astrocytes isolated from a sEH knockout (sEH−/−) background were also applied.

Results

The immunoreactivity of sEH was increased in activated astrocytes in parallel with the progression of AD in APP/PS1 mice. Our data from primary astrocyte cultures further demonstrate that the overexpression of sEH ameliorated, while the silencing of sEH mRNA enhanced, the lipopolysaccharides (LPS)-induced expression of pro-inflammatory markers, such as inducible nitric oxide, cyclooxygenase 2 (COX-2), and pro-inflammatory cytokines. These findings suggest that sEH negatively regulates astrocyte immune responses. Enhanced immune responses found in LPS-activated sEH−/− astrocytes also support the notion that the expression of sEH could suppress the immune responses during astrocyte activation. Similarly, sEH−/− mice that received intraperitoneal injection of LPS showed exacerbated astrocyte activation in the brain, as observed by the elevated expression of glial fibrillary acidic protein (GFAP) and pro-inflammatory markers. Moreover, our data show that the phosphorylation of the signal transducer and activator of transcription 3 (STAT3) was upregulated in activated astrocytes from sEH mouse brains, and the pharmacological blockade of STAT3 activity alleviated the pro-inflammatory effects of sEH deletion in LPS-activated primary astrocytes.

Conclusions

Our results provide evidence, for the first time, showing that sEH negatively regulates astrocytic immune responses and GFAP expression, while the underlying mechanism at least partly involves the downregulation of STAT3 phosphorylation. The discovery of a novel function for sEH in the negative control of astrocytic immune responses involving STAT3 activation confers further insights into the regulatory machinery of astrocyte activation during the development of neurodegeneration.
Appendix
Available only for authorised users
Literature
1.
go back to reference Li C, Zhao R, Gao K, Wei Z, Yin MY, Lau LT, Chui D, Yu AC. Astrocytes: implications for neuroinflammatory pathogenesis of Alzheimer’s disease. Curr Alzheimer Res. 2011;8:67–80.CrossRef Li C, Zhao R, Gao K, Wei Z, Yin MY, Lau LT, Chui D, Yu AC. Astrocytes: implications for neuroinflammatory pathogenesis of Alzheimer’s disease. Curr Alzheimer Res. 2011;8:67–80.CrossRef
2.
go back to reference Zhao J, O’Connor T, Vassar R. The contribution of activated astrocytes to Abeta production: implications for Alzheimer’s disease pathogenesis. J Neuroinflammation. 2011;8:150.CrossRef Zhao J, O’Connor T, Vassar R. The contribution of activated astrocytes to Abeta production: implications for Alzheimer’s disease pathogenesis. J Neuroinflammation. 2011;8:150.CrossRef
3.
go back to reference Aarum J, Sandberg K, Haeberlein SL, Persson MA. Migration and differentiation of neural precursor cells can be directed by microglia. Proc Natl Acad Sci U S A. 2003;100:15983–8.CrossRef Aarum J, Sandberg K, Haeberlein SL, Persson MA. Migration and differentiation of neural precursor cells can be directed by microglia. Proc Natl Acad Sci U S A. 2003;100:15983–8.CrossRef
4.
go back to reference Bilbo SD, Schwarz JM. Early-life programming of later-life brain and behavior: a critical role for the immune system. Front Behav Neurosci. 2009;3:14.CrossRef Bilbo SD, Schwarz JM. Early-life programming of later-life brain and behavior: a critical role for the immune system. Front Behav Neurosci. 2009;3:14.CrossRef
5.
go back to reference Ben Achour S, Pascual O. Glia: the many ways to modulate synaptic plasticity. Neurochem Int. 2010;57:440–5.CrossRef Ben Achour S, Pascual O. Glia: the many ways to modulate synaptic plasticity. Neurochem Int. 2010;57:440–5.CrossRef
6.
go back to reference Agostinho P, Cunha RA, Oliveira C. Neuroinflammation, oxidative stress and the pathogenesis of Alzheimer’s disease. Curr Pharm Des. 2010;16:2766–78.CrossRef Agostinho P, Cunha RA, Oliveira C. Neuroinflammation, oxidative stress and the pathogenesis of Alzheimer’s disease. Curr Pharm Des. 2010;16:2766–78.CrossRef
7.
go back to reference Verri M, Pastoris O, Dossena M, Aquilani R, Guerriero F, Cuzzoni G, Venturini L, Ricevuti G, Bongiorno AI. Mitochondrial alterations, oxidative stress and neuroinflammation in Alzheimer’s disease. Int J Immunopathol Pharmacol. 2012;25:345–53.CrossRef Verri M, Pastoris O, Dossena M, Aquilani R, Guerriero F, Cuzzoni G, Venturini L, Ricevuti G, Bongiorno AI. Mitochondrial alterations, oxidative stress and neuroinflammation in Alzheimer’s disease. Int J Immunopathol Pharmacol. 2012;25:345–53.CrossRef
8.
go back to reference Heneka MT, Carson MJ, El Khoury J, Landreth GE, Brosseron F, Feinstein DL, Jacobs AH, Wyss-Coray T, Vitorica J, Ransohoff RM, et al. Neuroinflammation in Alzheimer’s disease. Lancet Neurol. 2015;14:388–405.CrossRef Heneka MT, Carson MJ, El Khoury J, Landreth GE, Brosseron F, Feinstein DL, Jacobs AH, Wyss-Coray T, Vitorica J, Ransohoff RM, et al. Neuroinflammation in Alzheimer’s disease. Lancet Neurol. 2015;14:388–405.CrossRef
9.
go back to reference Bronzuoli MR, Iacomino A, Steardo L, Scuderi C. Targeting neuroinflammation in Alzheimer’s disease. J Inflamm Res. 2016;9:199–208.CrossRef Bronzuoli MR, Iacomino A, Steardo L, Scuderi C. Targeting neuroinflammation in Alzheimer’s disease. J Inflamm Res. 2016;9:199–208.CrossRef
10.
go back to reference O’Callaghan JP, Kelly KA, VanGilder RL, Sofroniew MV, Miller DB. Early activation of STAT3 regulates reactive astrogliosis induced by diverse forms of neurotoxicity. PLoS One. 2014;9:e102003.CrossRef O’Callaghan JP, Kelly KA, VanGilder RL, Sofroniew MV, Miller DB. Early activation of STAT3 regulates reactive astrogliosis induced by diverse forms of neurotoxicity. PLoS One. 2014;9:e102003.CrossRef
11.
go back to reference Peng H, Sun L, Jia B, Lan X, Zhu B, Wu Y, Zheng J. HIV-1-infected and immune-activated macrophages induce astrocytic differentiation of human cortical neural progenitor cells via the STAT3 pathway. PLoS One. 2011;6:e19439.CrossRef Peng H, Sun L, Jia B, Lan X, Zhu B, Wu Y, Zheng J. HIV-1-infected and immune-activated macrophages induce astrocytic differentiation of human cortical neural progenitor cells via the STAT3 pathway. PLoS One. 2011;6:e19439.CrossRef
12.
go back to reference Beurel E, Jope RS. Lipopolysaccharide-induced interleukin-6 production is controlled by glycogen synthase kinase-3 and STAT3 in the brain. J Neuroinflammation. 2009;6:9.CrossRef Beurel E, Jope RS. Lipopolysaccharide-induced interleukin-6 production is controlled by glycogen synthase kinase-3 and STAT3 in the brain. J Neuroinflammation. 2009;6:9.CrossRef
13.
go back to reference Fuller S, Munch G, Steele M. Activated astrocytes: a therapeutic target in Alzheimer’s disease? Expert Rev Neurother. 2009;9:1585–94.CrossRef Fuller S, Munch G, Steele M. Activated astrocytes: a therapeutic target in Alzheimer’s disease? Expert Rev Neurother. 2009;9:1585–94.CrossRef
14.
go back to reference Gonzalez-Reyes RE, Nava-Mesa MO, Vargas-Sanchez K, Ariza-Salamanca D, Mora-Munoz L. Involvement of astrocytes in Alzheimer’s disease from a neuroinflammatory and oxidative stress perspective. Front Mol Neurosci. 2017;10:427.CrossRef Gonzalez-Reyes RE, Nava-Mesa MO, Vargas-Sanchez K, Ariza-Salamanca D, Mora-Munoz L. Involvement of astrocytes in Alzheimer’s disease from a neuroinflammatory and oxidative stress perspective. Front Mol Neurosci. 2017;10:427.CrossRef
15.
go back to reference Zamanian JL, Xu L, Foo LC, Nouri N, Zhou L, Giffard RG, Barres BA. Genomic analysis of reactive astrogliosis. J Neurosci. 2012;32:6391–410.CrossRef Zamanian JL, Xu L, Foo LC, Nouri N, Zhou L, Giffard RG, Barres BA. Genomic analysis of reactive astrogliosis. J Neurosci. 2012;32:6391–410.CrossRef
16.
go back to reference Voskuhl RR, Peterson RS, Song B, Ao Y, Morales LB, Tiwari-Woodruff S, Sofroniew MV. Reactive astrocytes form scar-like perivascular barriers to leukocytes during adaptive immune inflammation of the CNS. J Neurosci. 2009;29:11511–22.CrossRef Voskuhl RR, Peterson RS, Song B, Ao Y, Morales LB, Tiwari-Woodruff S, Sofroniew MV. Reactive astrocytes form scar-like perivascular barriers to leukocytes during adaptive immune inflammation of the CNS. J Neurosci. 2009;29:11511–22.CrossRef
17.
go back to reference Myer DJ, Gurkoff GG, Lee SM, Hovda DA, Sofroniew MV. Essential protective roles of reactive astrocytes in traumatic brain injury. Brain. 2006;129:2761–72.CrossRef Myer DJ, Gurkoff GG, Lee SM, Hovda DA, Sofroniew MV. Essential protective roles of reactive astrocytes in traumatic brain injury. Brain. 2006;129:2761–72.CrossRef
18.
go back to reference Wyss-Coray T, Loike JD, Brionne TC, Lu E, Anankov R, Yan F, Silverstein SC, Husemann J. Adult mouse astrocytes degrade amyloid-beta in vitro and in situ. Nat Med. 2003;9:453–7.CrossRef Wyss-Coray T, Loike JD, Brionne TC, Lu E, Anankov R, Yan F, Silverstein SC, Husemann J. Adult mouse astrocytes degrade amyloid-beta in vitro and in situ. Nat Med. 2003;9:453–7.CrossRef
19.
go back to reference Vehmas AK, Kawas CH, Stewart WF, Troncoso JC. Immune reactive cells in senile plaques and cognitive decline in Alzheimer’s disease. Neurobiol Aging. 2003;24:321–31.CrossRef Vehmas AK, Kawas CH, Stewart WF, Troncoso JC. Immune reactive cells in senile plaques and cognitive decline in Alzheimer’s disease. Neurobiol Aging. 2003;24:321–31.CrossRef
20.
go back to reference Koistinaho M, Lin S, Wu X, Esterman M, Koger D, Hanson J, Higgs R, Liu F, Malkani S, Bales KR, Paul SM. Apolipoprotein E promotes astrocyte colocalization and degradation of deposited amyloid-beta peptides. Nat Med. 2004;10:719–26.CrossRef Koistinaho M, Lin S, Wu X, Esterman M, Koger D, Hanson J, Higgs R, Liu F, Malkani S, Bales KR, Paul SM. Apolipoprotein E promotes astrocyte colocalization and degradation of deposited amyloid-beta peptides. Nat Med. 2004;10:719–26.CrossRef
21.
go back to reference Chun H, Lee CJ. Reactive astrocytes in Alzheimer’s disease: a double-edged sword. Neurosci Res. 2018;126:44–52.CrossRef Chun H, Lee CJ. Reactive astrocytes in Alzheimer’s disease: a double-edged sword. Neurosci Res. 2018;126:44–52.CrossRef
22.
go back to reference Shie FS, Woltjer RL. Manipulation of microglial activation as a therapeutic strategy in Alzheimer’s disease. Curr Med Chem. 2007;14:2865–71.CrossRef Shie FS, Woltjer RL. Manipulation of microglial activation as a therapeutic strategy in Alzheimer’s disease. Curr Med Chem. 2007;14:2865–71.CrossRef
23.
go back to reference Simpkins AN, Rudic RD, Schreihofer DA, Roy S, Manhiani M, Tsai HJ, Hammock BD, Imig JD. Soluble epoxide inhibition is protective against cerebral ischemia via vascular and neural protection. Am J Pathol. 2009;174:2086–95.CrossRef Simpkins AN, Rudic RD, Schreihofer DA, Roy S, Manhiani M, Tsai HJ, Hammock BD, Imig JD. Soluble epoxide inhibition is protective against cerebral ischemia via vascular and neural protection. Am J Pathol. 2009;174:2086–95.CrossRef
24.
go back to reference Shaik JS, Ahmad M, Li W, Rose ME, Foley LM, Hitchens TK, Graham SH, Hwang SH, Hammock BD, Poloyac SM. Soluble epoxide hydrolase inhibitor trans-4-[4-(3-adamantan-1-yl-ureido)-cyclohexyloxy]-benzoic acid is neuroprotective in rat model of ischemic stroke. Am J Physiol Heart Circ Physiol. 2013;305:H1605–13.CrossRef Shaik JS, Ahmad M, Li W, Rose ME, Foley LM, Hitchens TK, Graham SH, Hwang SH, Hammock BD, Poloyac SM. Soluble epoxide hydrolase inhibitor trans-4-[4-(3-adamantan-1-yl-ureido)-cyclohexyloxy]-benzoic acid is neuroprotective in rat model of ischemic stroke. Am J Physiol Heart Circ Physiol. 2013;305:H1605–13.CrossRef
25.
go back to reference Zhang W, Otsuka T, Sugo N, Ardeshiri A, Alhadid YK, Iliff JJ, DeBarber AE, Koop DR, Alkayed NJ. Soluble epoxide hydrolase gene deletion is protective against experimental cerebral ischemia. Stroke. 2008;39:2073–8.CrossRef Zhang W, Otsuka T, Sugo N, Ardeshiri A, Alhadid YK, Iliff JJ, DeBarber AE, Koop DR, Alkayed NJ. Soluble epoxide hydrolase gene deletion is protective against experimental cerebral ischemia. Stroke. 2008;39:2073–8.CrossRef
26.
go back to reference Wu C-H, Shyue S-K, Hung T-H, Wen S, Lin C-C, Chang C-F, Chen S-F. Genetic deletion or pharmacological inhibition of soluble epoxide hydrolase reduces brain damage and attenuates neuroinflammation after intracerebral hemorrhage. J Neuroinflammation. 2017;14:230.CrossRef Wu C-H, Shyue S-K, Hung T-H, Wen S, Lin C-C, Chang C-F, Chen S-F. Genetic deletion or pharmacological inhibition of soluble epoxide hydrolase reduces brain damage and attenuates neuroinflammation after intracerebral hemorrhage. J Neuroinflammation. 2017;14:230.CrossRef
27.
go back to reference Tu R, Armstrong J, Lee KS, Hammock BD, Sapirstein A, Koehler RC. Soluble epoxide hydrolase inhibition decreases reperfusion injury after focal cerebral ischemia. Sci Rep. 2018;8:5279.CrossRef Tu R, Armstrong J, Lee KS, Hammock BD, Sapirstein A, Koehler RC. Soluble epoxide hydrolase inhibition decreases reperfusion injury after focal cerebral ischemia. Sci Rep. 2018;8:5279.CrossRef
28.
go back to reference Zarriello S, Tuazon JP, Corey S, Schimmel S, Rajani M, Gorsky A, Incontri D, Hammock BD, Borlongan CV. Humble beginnings with big goals: small molecule soluble epoxide hydrolase inhibitors for treating CNS disorders. Prog Neurobiol. 2019;172:23–39.CrossRef Zarriello S, Tuazon JP, Corey S, Schimmel S, Rajani M, Gorsky A, Incontri D, Hammock BD, Borlongan CV. Humble beginnings with big goals: small molecule soluble epoxide hydrolase inhibitors for treating CNS disorders. Prog Neurobiol. 2019;172:23–39.CrossRef
29.
go back to reference Panigrahy D, Greene ER, Pozzi A, Wang DW, Zeldin DC. EET signaling in cancer. Cancer Metastasis Rev. 2011;30:525–40.CrossRef Panigrahy D, Greene ER, Pozzi A, Wang DW, Zeldin DC. EET signaling in cancer. Cancer Metastasis Rev. 2011;30:525–40.CrossRef
30.
go back to reference Cronin A, Mowbray S, Durk H, Homburg S, Fleming I, Fisslthaler B, Oesch F, Arand M. The N-terminal domain of mammalian soluble epoxide hydrolase is a phosphatase. Proc Natl Acad Sci U S A. 2003;100:1552–7.CrossRef Cronin A, Mowbray S, Durk H, Homburg S, Fleming I, Fisslthaler B, Oesch F, Arand M. The N-terminal domain of mammalian soluble epoxide hydrolase is a phosphatase. Proc Natl Acad Sci U S A. 2003;100:1552–7.CrossRef
31.
go back to reference Enayetallah AE, French RA, Thibodeau MS, Grant DF. Distribution of soluble epoxide hydrolase and of cytochrome P450 2C8, 2C9, and 2 J2 in human tissues. J Histochem Cytochem. 2004;52:447–54.CrossRef Enayetallah AE, French RA, Thibodeau MS, Grant DF. Distribution of soluble epoxide hydrolase and of cytochrome P450 2C8, 2C9, and 2 J2 in human tissues. J Histochem Cytochem. 2004;52:447–54.CrossRef
32.
go back to reference Sura P, Sura R, Enayetallah AE, Grant DF. Distribution and expression of soluble epoxide hydrolase in human brain. J Histochem Cytochem. 2008;56:551–9.CrossRef Sura P, Sura R, Enayetallah AE, Grant DF. Distribution and expression of soluble epoxide hydrolase in human brain. J Histochem Cytochem. 2008;56:551–9.CrossRef
33.
go back to reference Hung YW, Hung SW, Wu YC, Wong LK, Lai MT, Shih YH, Lee TS, Lin YY. Soluble epoxide hydrolase activity regulates inflammatory responses and seizure generation in two mouse models of temporal lobe epilepsy. Brain Behav Immun. 2015;43:118–29.CrossRef Hung YW, Hung SW, Wu YC, Wong LK, Lai MT, Shih YH, Lee TS, Lin YY. Soluble epoxide hydrolase activity regulates inflammatory responses and seizure generation in two mouse models of temporal lobe epilepsy. Brain Behav Immun. 2015;43:118–29.CrossRef
34.
go back to reference Maragakis NJ, Rothstein JD. Mechanisms of disease: astrocytes in neurodegenerative disease. Nat Clin Pract Neurol. 2006;2:679–89.CrossRef Maragakis NJ, Rothstein JD. Mechanisms of disease: astrocytes in neurodegenerative disease. Nat Clin Pract Neurol. 2006;2:679–89.CrossRef
35.
go back to reference Yamanaka K, Chun SJ, Boillee S, Fujimori-Tonou N, Yamashita H, Gutmann DH, Takahashi R, Misawa H, Cleveland DW. Astrocytes as determinants of disease progression in inherited amyotrophic lateral sclerosis. Nat Neurosci. 2008;11:251–3.CrossRef Yamanaka K, Chun SJ, Boillee S, Fujimori-Tonou N, Yamashita H, Gutmann DH, Takahashi R, Misawa H, Cleveland DW. Astrocytes as determinants of disease progression in inherited amyotrophic lateral sclerosis. Nat Neurosci. 2008;11:251–3.CrossRef
36.
go back to reference Anderson MA, Ao Y, Sofroniew MV. Heterogeneity of reactive astrocytes. Neurosci Lett. 2014;565:23–9.CrossRef Anderson MA, Ao Y, Sofroniew MV. Heterogeneity of reactive astrocytes. Neurosci Lett. 2014;565:23–9.CrossRef
37.
go back to reference Mathur R, Ince PG, Minett T, Garwood CJ, Shaw PJ, Matthews FE, Brayne C, Simpson JE, Wharton SB, Function MRCC, Ageing Neuropathology Study G. A reduced astrocyte response to beta-amyloid plaques in the ageing brain associates with cognitive impairment. PLoS One. 2015;10:e0118463.CrossRef Mathur R, Ince PG, Minett T, Garwood CJ, Shaw PJ, Matthews FE, Brayne C, Simpson JE, Wharton SB, Function MRCC, Ageing Neuropathology Study G. A reduced astrocyte response to beta-amyloid plaques in the ageing brain associates with cognitive impairment. PLoS One. 2015;10:e0118463.CrossRef
38.
go back to reference Wyss-Coray T, Mucke L. Inflammation in neurodegenerative disease--a double-edged sword. Neuron. 2002;35:419–32.CrossRef Wyss-Coray T, Mucke L. Inflammation in neurodegenerative disease--a double-edged sword. Neuron. 2002;35:419–32.CrossRef
39.
go back to reference Murray PJ. The primary mechanism of the IL-10-regulated antiinflammatory response is to selectively inhibit transcription. Proc Natl Acad Sci U S A. 2005;102:8686–91.CrossRef Murray PJ. The primary mechanism of the IL-10-regulated antiinflammatory response is to selectively inhibit transcription. Proc Natl Acad Sci U S A. 2005;102:8686–91.CrossRef
40.
go back to reference Hutchins AP, Diez D, Miranda-Saavedra D. The IL-10/STAT3-mediated anti-inflammatory response: recent developments and future challenges. Brief Funct Genomics. 2013;12:489–98.CrossRef Hutchins AP, Diez D, Miranda-Saavedra D. The IL-10/STAT3-mediated anti-inflammatory response: recent developments and future challenges. Brief Funct Genomics. 2013;12:489–98.CrossRef
Metadata
Title
Soluble epoxide hydrolase modulates immune responses in activated astrocytes involving regulation of STAT3 activity
Authors
Chia-Chi Hung
Yi-Hsuan Lee
Yi-Min Kuo
Pei-Chien Hsu
Huey-Jen Tsay
Ying-Ting Hsu
Chih-Chin Lee
Jia-Jun Liang
Feng-Shiun Shie
Publication date
01-12-2019
Publisher
BioMed Central
Published in
Journal of Neuroinflammation / Issue 1/2019
Electronic ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-019-1508-2

Other articles of this Issue 1/2019

Journal of Neuroinflammation 1/2019 Go to the issue