Skip to main content
Top
Published in: BMC Neurology 1/2019

Open Access 01-12-2019 | Stroke | Research article

PPAR-γ promotes p38 MAP kinase-mediated endothelial cell permeability through activating Sirt3

Authors: Zhenzhen Zhao, Xiaoxiu Zhang, Yuanqiang Dai, Ke Pan, Yu Deng, Yan Meng, Tao Xu

Published in: BMC Neurology | Issue 1/2019

Login to get access

Abstract

Background

Ischemia-reperfusion (I/R)-induced vascular dysfunction is the main factor to acute ischemic stroke. Sirt3 is one of the sirtuin family members, which plays an important role in the development of neurological diseases.

Methods

In this study, we constructed I/R injury model on HBMEC cells and induced the overexpression of Sirt3 in model cells. Meanwhile, the p38 activator U-46619 was used to examine the connection between Sirt3 and p38. We also examined the level of endothelial associated proteins, including occluding, ZO-1 and claudin-4 by using qRT-PCR and western blot.

Results

Our findings indicated that overexpression of Sirt3 decreased the permeability of model cells and promoted in the growth of endothelial cells. However, the activation of p38 could antagonize the function of Sirt3 in HBMEC cells. Moreover, Our results indicated a positive correlation between Sirt3 and inter-endothelial junction proteins. Importantly, PPAR-γ agonist and inhibitor were utilized to investigate the role of PPAR-γ in Sirt3 mediated cell function. Sirt3 was targeted by PPAR-γ in model cells.

Conclusions

Taken together, this research not only demonstrated PPAR-γ might benefit to the growth of endothelial cell though activating Sirt3 but also indicated its potential value in the treatment for ischemic stroke.
Literature
1.
go back to reference Zhang, Y., et al., Zhang Y, Zhang X, Park TS, Gidday JM Cerebral endothelial cell apoptosis after ischemia-reperfusion-role of PARP activation and AIF translocation. J Cereb Blood Flow Metab 25:868–877. 2005. 25(7): p. 868–877.CrossRef Zhang, Y., et al., Zhang Y, Zhang X, Park TS, Gidday JM Cerebral endothelial cell apoptosis after ischemia-reperfusion-role of PARP activation and AIF translocation. J Cereb Blood Flow Metab 25:868–877. 2005. 25(7): p. 868–877.CrossRef
2.
go back to reference Pasban E, Panahpour H, Vahdati A. Early oxygen therapy does not protect the brain from vasogenic edema following acute ischemic stroke in adult male rats. Sci Rep. 2017;7(1):3221.CrossRefPubMedPubMedCentral Pasban E, Panahpour H, Vahdati A. Early oxygen therapy does not protect the brain from vasogenic edema following acute ischemic stroke in adult male rats. Sci Rep. 2017;7(1):3221.CrossRefPubMedPubMedCentral
3.
go back to reference Furihata, T., et al., Hydrocortisone enhances the barrier properties of HBMEC/ciβ, a brain microvascular endothelial cell line, through mesenchymal-to-endothelial transition-like effects. Fluids and Barriers of the CNS,12,1(2015-03-05), 2015. 12(1): p. 7. Furihata, T., et al., Hydrocortisone enhances the barrier properties of HBMEC/ciβ, a brain microvascular endothelial cell line, through mesenchymal-to-endothelial transition-like effects. Fluids and Barriers of the CNS,12,1(2015-03-05), 2015. 12(1): p. 7.
5.
8.
go back to reference Naia L, Rego AC. Sirtuins: double players in Huntington's disease. BBA - Molecular Basis of Disease. 2015;1852(10):2183–94.CrossRefPubMed Naia L, Rego AC. Sirtuins: double players in Huntington's disease. BBA - Molecular Basis of Disease. 2015;1852(10):2183–94.CrossRefPubMed
9.
go back to reference Dai SH, et al. Sirt3 confers protection against neuronal ischemia by inducing autophagy: involvement of the AMPK-mTOR pathway. Free Radic Biol Med. 2017;108:345.CrossRefPubMed Dai SH, et al. Sirt3 confers protection against neuronal ischemia by inducing autophagy: involvement of the AMPK-mTOR pathway. Free Radic Biol Med. 2017;108:345.CrossRefPubMed
10.
go back to reference Shioda N, Han F, Fukunaga K. Role of Akt and ERK signaling in the neurogenesis following brain ischemia. Int Rev Neurobiol. 2009;85:375–87.CrossRefPubMed Shioda N, Han F, Fukunaga K. Role of Akt and ERK signaling in the neurogenesis following brain ischemia. Int Rev Neurobiol. 2009;85:375–87.CrossRefPubMed
12.
go back to reference Sun W, et al. SIRT3: a new regulator of cardiovascular diseases. Oxidative Med Cell Longev. 2018;2018:7293861. Sun W, et al. SIRT3: a new regulator of cardiovascular diseases. Oxidative Med Cell Longev. 2018;2018:7293861.
13.
go back to reference Victor NA, et al. Altered PPARgamma expression and activation after transient focal ischemia in rats. Eur J Neurosci. 2006;24(6):1653–63.CrossRefPubMed Victor NA, et al. Altered PPARgamma expression and activation after transient focal ischemia in rats. Eur J Neurosci. 2006;24(6):1653–63.CrossRefPubMed
14.
go back to reference Chen YC, et al. Peroxisome proliferator-activated receptor gamma (PPAR-γ) and neurodegenerative disorders. Mol Neurobiol. 2012;46(1):114–24.CrossRefPubMed Chen YC, et al. Peroxisome proliferator-activated receptor gamma (PPAR-γ) and neurodegenerative disorders. Mol Neurobiol. 2012;46(1):114–24.CrossRefPubMed
15.
go back to reference Wu JS, et al. Clinacanthus nutans mitigates neuronal apoptosis and ischemic brain damage through augmenting the C/EBPβ-driven PPAR-γ transcription. Mol Neurobiol. 2017;2:1–14. Wu JS, et al. Clinacanthus nutans mitigates neuronal apoptosis and ischemic brain damage through augmenting the C/EBPβ-driven PPAR-γ transcription. Mol Neurobiol. 2017;2:1–14.
16.
go back to reference Wu JS, et al. PPAR-γ ameliorates neuronal apoptosis and ischemic brain injury via suppressing NF-κB-driven p22phox transcription. Mol Neurobiol. 2016;53(6):3626–45.CrossRefPubMed Wu JS, et al. PPAR-γ ameliorates neuronal apoptosis and ischemic brain injury via suppressing NF-κB-driven p22phox transcription. Mol Neurobiol. 2016;53(6):3626–45.CrossRefPubMed
17.
go back to reference Huang T, et al. D-allose protects the blood brain barrier through PPARγ-mediated anti-inflammatory pathway in the mice model of ischemia reperfusion injury. Brain Res. 2016;1642:478–86.CrossRefPubMed Huang T, et al. D-allose protects the blood brain barrier through PPARγ-mediated anti-inflammatory pathway in the mice model of ischemia reperfusion injury. Brain Res. 2016;1642:478–86.CrossRefPubMed
18.
19.
go back to reference Liu R, et al. XQ-1H protects against ischemic stroke by regulating microglia polarization through PPARγ pathway in mice. Int Immunopharmacol. 2018;57:72–81.CrossRefPubMed Liu R, et al. XQ-1H protects against ischemic stroke by regulating microglia polarization through PPARγ pathway in mice. Int Immunopharmacol. 2018;57:72–81.CrossRefPubMed
20.
go back to reference F, Y., et al., SIRT3 inhibits Ang II-induced transdifferentiation of cardiac fibroblasts through β-catenin/PPAR-γ signaling. Life Sciences, 2017. 186(16): p. 111. F, Y., et al., SIRT3 inhibits Ang II-induced transdifferentiation of cardiac fibroblasts through β-catenin/PPAR-γ signaling. Life Sciences, 2017. 186(16): p. 111.
21.
go back to reference Banno A, et al. PPARs: key regulators of airway inflammation and potential therapeutic targets in asthma. Nucl Receptor Res. 2018;5. Banno A, et al. PPARs: key regulators of airway inflammation and potential therapeutic targets in asthma. Nucl Receptor Res. 2018;5.
22.
go back to reference Collino M, Patel NS, Thiemermann C. PPARs as new therapeutic targets for the treatment of cerebral ischemia/reperfusion injury. Ther Adv Cardiovasc Dis. 2008;2(3):179–97.CrossRefPubMed Collino M, Patel NS, Thiemermann C. PPARs as new therapeutic targets for the treatment of cerebral ischemia/reperfusion injury. Ther Adv Cardiovasc Dis. 2008;2(3):179–97.CrossRefPubMed
23.
go back to reference Shi Y, et al. Rapid endothelial cytoskeletal reorganization enables early blood-brain barrier disruption and long-term ischaemic reperfusion brain injury. Nat Commun. 2016;7:10523.CrossRefPubMedPubMedCentral Shi Y, et al. Rapid endothelial cytoskeletal reorganization enables early blood-brain barrier disruption and long-term ischaemic reperfusion brain injury. Nat Commun. 2016;7:10523.CrossRefPubMedPubMedCentral
24.
go back to reference Zhang W, et al. Increased expression of bioactive chemokines in human cerebromicrovascular endothelial cells and astrocytes subjected to simulated ischemia in vitro. J Neuroimmunol. 1999;101(2):148–60.CrossRefPubMed Zhang W, et al. Increased expression of bioactive chemokines in human cerebromicrovascular endothelial cells and astrocytes subjected to simulated ischemia in vitro. J Neuroimmunol. 1999;101(2):148–60.CrossRefPubMed
25.
go back to reference Walsh TG, et al. Stabilization of brain microvascular endothelial barrier function by shear stress involves VE-cadherin signaling leading to modulation of pTyr-occludin levels. J Cell Physiol. 2011;226(11):3053–63.CrossRefPubMed Walsh TG, et al. Stabilization of brain microvascular endothelial barrier function by shear stress involves VE-cadherin signaling leading to modulation of pTyr-occludin levels. J Cell Physiol. 2011;226(11):3053–63.CrossRefPubMed
27.
go back to reference Birgit O, Richard D, Richard R. M, Development, maintenance and disruption of the blood-brain barrier. Nat Med. 2013;19(12):1584–96.CrossRef Birgit O, Richard D, Richard R. M, Development, maintenance and disruption of the blood-brain barrier. Nat Med. 2013;19(12):1584–96.CrossRef
28.
go back to reference Mcdonnell E, et al. SIRT3 regulates progression and development of diseases of aging. Trends in Endocrinology & Metabolism. 2015;26(9):486.CrossRef Mcdonnell E, et al. SIRT3 regulates progression and development of diseases of aging. Trends in Endocrinology & Metabolism. 2015;26(9):486.CrossRef
29.
go back to reference Nito C, et al. Role of the p38 mitogen-activated protein kinase/cytosolic phospholipase A2 signaling pathway in blood-brain barrier disruption after focal cerebral ischemia and reperfusion. Journal of Cerebral Blood Flow & Metabolism Official Journal of the International Society of Cerebral Blood Flow & Metabolism. 2008;28(10):1686.CrossRef Nito C, et al. Role of the p38 mitogen-activated protein kinase/cytosolic phospholipase A2 signaling pathway in blood-brain barrier disruption after focal cerebral ischemia and reperfusion. Journal of Cerebral Blood Flow & Metabolism Official Journal of the International Society of Cerebral Blood Flow & Metabolism. 2008;28(10):1686.CrossRef
30.
go back to reference Förster C. Tight junctions and the modulation of barrier function in disease. Histochemistry & Cell Biology. 2008;130(1):55–70.CrossRef Förster C. Tight junctions and the modulation of barrier function in disease. Histochemistry & Cell Biology. 2008;130(1):55–70.CrossRef
31.
go back to reference Cao C, et al. S1PR2 antagonist alleviates oxidative stress-enhanced brain endothelial permeability by attenuating p38 and Erk1/2-dependent cPLA2 phosphorylation. Cell Signal. 2018;53:151–61.CrossRefPubMed Cao C, et al. S1PR2 antagonist alleviates oxidative stress-enhanced brain endothelial permeability by attenuating p38 and Erk1/2-dependent cPLA2 phosphorylation. Cell Signal. 2018;53:151–61.CrossRefPubMed
32.
go back to reference Gao Z, et al. SIRT1 mediates Sphk1/S1P-induced proliferation and migration of endothelial cells. Int J Biochem Cell Biol. 2016;74:152–60.CrossRefPubMed Gao Z, et al. SIRT1 mediates Sphk1/S1P-induced proliferation and migration of endothelial cells. Int J Biochem Cell Biol. 2016;74:152–60.CrossRefPubMed
34.
go back to reference Wu JS, et al. Clinacanthus nutans mitigates neuronal apoptosis and ischemic brain damage through augmenting the C/EBPbeta-driven PPAR-gamma transcription. Mol Neurobiol. 2018;55(7):5425–38.CrossRefPubMed Wu JS, et al. Clinacanthus nutans mitigates neuronal apoptosis and ischemic brain damage through augmenting the C/EBPbeta-driven PPAR-gamma transcription. Mol Neurobiol. 2018;55(7):5425–38.CrossRefPubMed
35.
go back to reference Verma R, et al. Evaluation of the Neuroprotective effect of Sirt3 in experimental stroke. Transl Stroke Res. 2019;10(1):57–66.CrossRefPubMed Verma R, et al. Evaluation of the Neuroprotective effect of Sirt3 in experimental stroke. Transl Stroke Res. 2019;10(1):57–66.CrossRefPubMed
36.
go back to reference Culman J, et al. PPAR-gamma: therapeutic target for ischemic stroke. Trends Pharmacol Sci. 2007;28(5):244–9.CrossRefPubMed Culman J, et al. PPAR-gamma: therapeutic target for ischemic stroke. Trends Pharmacol Sci. 2007;28(5):244–9.CrossRefPubMed
37.
go back to reference Agarwal S, Yadav A, Chaturvedi RK. Peroxisome proliferator-activated receptors (PPARs) as therapeutic target in neurodegenerative disorders. Biochem Biophys Res Commun. 2017;483(4):1166–77.CrossRefPubMed Agarwal S, Yadav A, Chaturvedi RK. Peroxisome proliferator-activated receptors (PPARs) as therapeutic target in neurodegenerative disorders. Biochem Biophys Res Commun. 2017;483(4):1166–77.CrossRefPubMed
Metadata
Title
PPAR-γ promotes p38 MAP kinase-mediated endothelial cell permeability through activating Sirt3
Authors
Zhenzhen Zhao
Xiaoxiu Zhang
Yuanqiang Dai
Ke Pan
Yu Deng
Yan Meng
Tao Xu
Publication date
01-12-2019
Publisher
BioMed Central
Keyword
Stroke
Published in
BMC Neurology / Issue 1/2019
Electronic ISSN: 1471-2377
DOI
https://doi.org/10.1186/s12883-019-1508-y

Other articles of this Issue 1/2019

BMC Neurology 1/2019 Go to the issue