Mild Therapeutic Hypothermia Protects the Brain from Ischemia/Reperfusion Injury through Upregulation of iASPP

Xiangrong Liu, Shaohong Wen, Shunying Zhao, Feng Yan, Shangfeng Zhao, Di Wu, Xunming Ji, Xiangrong Liu, Shaohong Wen, Shunying Zhao, Feng Yan, Shangfeng Zhao, Di Wu, Xunming Ji

Abstract

Mild therapeutic hypothermia, a robust neuroprotectant, reduces neuronal apoptosis, but the precise mechanism is not well understood. Our previous study showed that a novel inhibitor of an apoptosis-stimulating protein of p53 (iASPP) might be involved in neuronal death after stroke. The aim of this study was to confirm the role of iASPP after stroke treated with mild therapeutic hypothermia. To address this, we mimicked ischemia/reperfusion injury in vitro by using oxygen-glucose deprivation/reperfusion (OGD/R) in primary rat neurons. In our in vivo approach, we induced middle cerebral artery occlusion (MCAO) for 60 min in C57/B6 mice. From the beginning of ischemia, focal mild hypothermia was applied for two hours. To evaluate the role of iASPP, small interfering RNA (siRNA) was injected intracerebroventricularly. Our results showed that mild therapeutic hypothermia increased the expression of iASPP and decreased the expression of its targets, Puma and Bax, and an apoptosis marker, cleaved caspase-3, in primary neurons under OGD/R. Increased iASPP expression and decreased ASPP1/2 expression were observed under hypothermia treatment in MCAO mice. iASPP siRNA (iASPPi) or hypothermia plus iASPPi application increased infarct volume, apoptosis and aggravated the neurological deficits in MCAO mice. Furthermore, iASPPi downregulated iASPP expression, and upregulated the expression of proapoptotic effectors, Puma, Bax and cleaved caspase-3, in mice after stroke treated with mild therapeutic hypothermia. In conclusion, mild therapeutic hypothermia protects against ischemia/reperfusion brain injury in mice by upregulating iASPP and thus attenuating apoptosis. iASPP may be a potential target in the therapy of stroke.

Keywords: apoptosis; hypothermia; iASPP protein; small interfering RNA; stroke; therapeutic.

Conflict of interest statement

Disclosure The authors have no conflicts to disclose about this work.

Figures

Figure 1.
Figure 1.
Mild therapeutic hypothermia regulates the expression of iASPP and its targets in primary neurons treated with OGD/R. (A) Representative protein in bands of iASPP, Puma, Bax and cleaved caspase-3 at 24 h after OGD from the Western blot. Actin served as a loading control. (B) Densitometric quantification of iASPP, Puma, Bax and cleaved caspase-3 from the Western blot, n = 4 per group. (C) Representative images showing expression of iASPP (green) in primary neurons (red) treated with OGD at 37°C or 33°C. DAPI was used as a nuclear marker. Bar, 50 μm.*p < 0.05, **p < 0.01, ***p < 0.001, by one-way ANOVA and Tukey’s test.
Figure 2.
Figure 2.
Mild therapeutic hypothermia augmented expression of iASPP after stroke. (A) Representative protein bands of iASPP from the Western blot. Actin served as a loading control. Mice subjected to normothermic or mild hypothermic stroke were euthanized at 4, 12, 24 and 72 h later. (B) Densitometric quantification of iASPP from the Western blot. *p < 0.05, ***p < 0.001, vs sham group under normothermia; # p < 0.05, ### p < 0.001, vs sham group under mild therapeutic hypothermia; ♦♦ p < 0.01, ♦♦♦ p < 0.01, vs corresponding groups under normothermia, by two-way ANOVA and Sidak’s test, n = 4 per group. (C) Representative images showing localization of iASPP (red) in neurons (green) of the cortex at 24 h after stroke. Bar, 50 μm.
Figure 3.
Figure 3.
Mild therapeutic hypothermia decreased expression of ASPP1 and ASPP2 after stroke. (A) Representative protein bands of ASPP1 and ASPP2 from the Western blot. Actin served as a loading control. Mice subjected to normothermic or mild hypothermic stroke were euthanized at 4, 12, 24 and 72 h later. (B) Densitometric quantification of ASPP1 and ASPP2 in Western blot, n = 4 per group. Data are expressed as mean ± SEM. *p < 0.05, **p < 0.01, vs sham group under normothermia; # p < 0.05 vs sham group under mild therapeutic hypothermia; ♦ p < 0.05, vs. corresponding group under normothermia, by two-way ANOVA and Sidak’s test. (C) Representative images showing expression of ASPP1 and ASPP2 (red) in neurons (green) of the cortex at 12 h after cerebral ischemia. DAPI was used as a nuclear marker.
Figure 4.
Figure 4.
Effect of iASPPi and iASPPi plus hypothermia on mice cerebral ischemic injury. (A) Representative brain slices with infarcts stained by 2, 3, 5-triphenyltetrazolium chloride from each group at 24 h after reperfusion. (B) Statistical analysis of the percentage of infarct volume in different treated groups, n = 10-12 per group. (C) Statistical analysis of neurological severity scores (NSS) in different treated groups, n = 13-16 per group. (D) Representative TUNEL staining (green) of brain slices from different groups at 24 h after reperfusion. DAPI was used as a nuclear marker. (E) Statistical analysis of apoptosis cells showing that mild hypothermia decreased cell apoptosis, n = 4-6 per group. 37°C, normothermia. 33°C, mild therapeutic hypothermia. Small interfering RNA (siRNA) was intracerebroventricularly injected. Coni, control siRNA; iASPP siRNA (iASPPi). Data are expressed as mean ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001, by one-way ANOVA and Tukey’s test.
Figure 5.
Figure 5.
Effect of iASPPi and iASPPi plus hypothermia on the expression of iASPP and its targets in MCAO mice. (A) Representative protein in bands of iASPP, Puma, Bax and cleaved caspase-3 at 24 h after cerebral ischemia from the Western blot. Actin served as a loading control. (B) Densitometric quantification of iASPP, Puma, Bax and cleaved caspase-3 measured by Western blot. C-casp3, cleaved caspase-3, n = 4-6 per group. 37°C, normothermia. 33°C, mild therapeutic hypothermia. Small interfering RNA (siRNA) was intracerebroventricularly injected. Coni, control siRNA; iASPP siRNA (iASPPi). Data are expressed as mean ± SEM. N.S., not significant. *p < 0.05, **p < 0.01, ***p < 0.001, by one-way ANOVA and Tukey’s test.

References

    1. Lahr MM, Luijckx GJ, Vroomen PC, van der Zee DJ, Buskens E (2013). The chain of care enabling tPA treatment in acute ischemic stroke: a comprehensive review of organisational models. J Neurol, 260: 960-968.
    1. Groysman LI, Emanuel BA, Kim-Tenser MA, Sung GY, Mack WJ (2011). Therapeutic hypothermia in acute ischemic stroke. Neurosurg Focus, 30: E17.
    1. Choi HA, Badjatia N, Mayer SA (2012). Hypothermia for acute brain injury--mechanisms and practical aspects. Nat Rev Neurol, 8: 214-222.
    1. Darwazeh R, Yan Y (2013). Mild hypothermia as a treatment for central nervous system injuries: Positive or negative effects. Neural Regen Res, 8: 2677-2686.
    1. Han Z, Liu X, Luo Y, Ji X (2015). Therapeutic hypothermia for stroke: Where to go? Exp Neurol, 272: 67-77.
    1. Kim JY, Yenari MA (2015). Hypothermia for treatment of stroke. Brain Circ, 1:14-25.
    1. Yenari MA, Han HS (2012). Neuroprotective mechanisms of hypothermia in brain ischaemia. Nat Rev Neurosci, 13:267-278.
    1. Alsafadi S, Tourpin S, André F, Vassal G, Ahomadegbe JC (2009). P53 family: at the crossroads in cancer therapy. Curr Med Chem, 16:4328-4344.
    1. Fortin A, Cregan SP, MacLaurin JG, Kushwaha N, Hickman ES, Thompson CS, et al. (2001). APAF1 is a key transcriptional target for p53 in the regulation of neuronal cell death. J Cell Biol, 155:207-216.
    1. Pei L, Shang Y, Jin H, Wang S, Wei N, Yan H, et al. (2014). DAPK1-p53 interaction converges necrotic and apoptotic pathways of ischemic neuronal death. J Neurosci, 34:6546-6556.
    1. Hong LZ, Zhao XY, Zhang HL (2010). p53-mediated neuronal cell death in ischemic brain injury. Neurosci Bull, 26:232-240.
    1. Di Giovanni S, Rathore K (2012). p53-Dependent pathways in neurite outgrowth and axonal regeneration. Cell Tissue Res, 349:87-95.
    1. Sullivan A, Lu X (2007). ASPP: a new family of oncogenes and tumour suppressor genes. Br J Cancer, 96:196-200.
    1. Robinson RA, Lu X, Jones EY, Siebold C (2008). Biochemical and structural studies of ASPP proteins reveal differential binding to p53, p63, and p73. Structure, 16:259-268.
    1. Cai Y, Qiu S, Gao X, Gu SZ, Liu ZJ (2012). iASPP inhibits p53-independent apoptosis by inhibiting transcriptional activity of p63/p73 on promoters of proapoptotic genes. Apoptosis, 17:777-783.
    1. Wilson AM, Chiodo VA, Boye SL, Brecha NC, Hauswirth WW, Di Polo A (2014). Inhibitor of apoptosis-stimulating protein of p53 (iASPP) is required for neuronal survival after axonal injury. PLoS One, 9: e94175.
    1. Liu X, Li F, Zhao S, Luo Y, Kang J, Zhao H, et al. (2013). MicroRNA-124-mediated regulation of inhibitory member of apoptosis-stimulating protein of p53 family in experimental stroke. Stroke, 44: 1973-1980.
    1. Cao G, Pei W, Ge H, Liang Q, Luo Y, Sharp FR, et al. (2002). In vivo delivery of a Bcl-xl fusion protein containing the TAT protein transduction domain protects against ischemic brain injury and neuronal apoptosis. J Neurosci, 22: 5423-5431.
    1. Sawada M, Alkayed NJ, Goto S, Crain BJ, Traystman RJ, Shaivitz A, et al. (2000). Estrogen receptor antagonist ICI182,780 exacerbates ischemic injury in female mouse. J Cereb Blood Flow Metab, 20:112-118.
    1. Stahel PF, Shohami E, Younis FM, Kariya K, Otto VI, Lenzlinger PM, et al. (2000). Experimental closed head injury: analysis of neurological outcome, blood-brain barrier dysfunction, intracranial neutrophil infiltration, and neuronal cell death in mice deficient in genes for pro-inflammatory cytokines. J Cereb Blood Flow Metab, 20:369-380.
    1. Chen J, Li Y, Wang L, Zhang Z, Lu D, Lu M, et al. (2001). Therapeutic benefit of intravenous administration of bone marrow stromal cells after cerebral ischemia in rats. Stroke, 32:1005-1011.
    1. Bindra CS, Jaggi AS, Singh N (2014). Role of P2X7 purinoceptors in neuroprotective mechanism of ischemic postconditioning in mice. Mol Cell Biochem, 390:161-173.
    1. Xiong X, Barreto GE, Xu L, Ouyang YB, Xie X, Giffard RG (2011). Increased brain injury and worsened neurological outcome in interleukin-4 knockout mice after transient focal cerebral ischemia. Stroke, 42:2026-2032.
    1. Liu X, Zhao S, Liu F, Kang J, Xiao A, Li F, et al. (2014). Remote ischemic postconditioning alleviates cerebral ischemic injury by attenuating endoplasmic reticulum stress-mediated apoptosis. Transl Stroke Res, 5:692-700.
    1. Mazumder S, Plesca D, Almasan A (2008). Caspase-3 activation is a critical determinant of genotoxic stress-induced apoptosis. Methods Mol Biol, 414:13-21.
    1. Hu X, Li P, Guo Y, Wang H, Leak RK, Chen S, et al. (2012). Microglia/macrophage polarization dynamics reveal novel mechanism of injury expansion after focal cerebral ischemia. Stroke, 43:3063-3070.
    1. Silasi G, Colbourne F (2011). Therapeutic hypothermia influences cell genesis and survival in the rat hippocampus following global ischemia. J Cereb Blood Flow Metab, 31:1725-1735.
    1. Bergamaschi D, Samuels Y, O’Neil NJ, Trigiante G, Crook T, Hsieh JK, et al. (2003). iASPP oncoprotein is a key inhibitor of p53 conserved from worm to human. Nat Genet, 33:162-167.
    1. Li G, Wang R, Gao J, Deng K, Wei J, Wei Y (2011). RNA interference-mediated silencing of iASPP induces cell proliferation inhibition and G0/G1 cell cycle arrest in U251 human glioblastoma cells. Mol Cell Biochem, 350:193-200.
    1. Liu H, Wang M, Diao S, Rao Q, Zhang X, Xing H, et al. (2009). siRNA-mediated down-regulation of iASPP promotes apoptosis induced by etoposide and daunorubicin in leukemia cells expressing wild-type p53. Leuk Res, 33:1243-1248.
    1. Zhang X, Wang M, Zhou C, Chen S, Wang J (2005). The expression of iASPP in acute leukemias. Leuk Res, 29:179-183.
    1. Liu X, Kang J, Liu F, Wen S, Zeng X, Liu K, et al. (2016). Overexpression of iASPP-SV in glioma is associated with poor prognosis by promoting cell viability and antagonizing apoptosis. Tumour Biol, 37:6323-6330.
    1. Kong F, Shi X, Li H, Li P, Yu J, Li X, et al. (2015). Increased expression of iASPP correlates with poor prognosis in FIGO IA2-IIA cervical adenocarcinoma following a curative resection. Am J Cancer Res, 5:1217-1224.
    1. Jia Y, Peng L, Rao Q, Xing H, Huai L, Yu P, et al. (2014). Oncogene iASPP enhances self-renewal of hematopoietic stem cells and facilitates their resistance to chemotherapy and irradiation. FASEB J, 28: 2816-2827.
    1. Yi H, Huang Y, Yang F, Liu W, He S, Hu X (2017). MicroRNA-182 aggravates cerebral ischemia injury by targeting inhibitory member of the ASPP family (iASPP). Arch Biochem Biophys, 620: 52-58.
    1. Sun J, Ji J, Huo G, Song Q, Zhang X (2015). miR-182 induces cervical cancer cell apoptosis through inhibiting the expression of DNMT3a. Int J Clin Exp Pathol, 8:4755-4763.
    1. Hu Y, Ge W, Wang X, Sutendra G, Zhao K, Dedeić Z, et al. (2015). Caspase cleavage of iASPP potentiates its ability to inhibit p53 and NF-κB. Oncotarget, 6:42478-42490.
    1. Dong P, Ihira K, Hamada J, Watari H, Yamada T, Hosaka M, et al. (2015). Reactivating p53 functions by suppressing its novel inhibitor iASPP: a potential therapeutic opportunity in p53 wild-type tumors. Oncotarget, 6:19968-19975.
    1. Chipuk JE, Green DR. Dissecting p53-dependent apoptosis (2006). Cell Death Differ, 13:994-1002.
    1. Zhang H, Xu G, Zhang J, Murong S, Mei Y, Tong E (2010). Mild hypothermia reduces ischemic neuron death via altering the expression of p53 and bcl-2. Neurol Res, 32: 384-389.
    1. Tomasevic G, Kamme F, Stubberöd P, Wieloch M, Wieloch T (1999). The tumor suppressor p53 and its response gene p21WAF1/Cip1 are not markers of neuronal death following transient global cerebral ischemia. Neuroscience, 90: 781-792.
    1. Bui T, Sequeira J, Wen TC, Sola A, Higashi Y, Kondoh H, et al. (2009). ZEB1 links p63 and p73 in a novel neuronal survival pathway rapidly induced in response to cortical ischemia. PLoS One, 4:e4373.
    1. Pozniak CD, Radinovic S, Yang A, McKeon F, Kaplan DR, Miller FD (2000). An anti-apoptotic role for the p53 family member, p73, during developmental neuron death. Science, 289:304-306.
    1. Pozniak CD, Barnabé-Heider F, Rymar VV, Lee AF, Sadikot AF, Miller FD (2002). p73 is required for survival and maintenance of CNS neurons. J Neurosci, 22:9800-9809.
    1. Melino G, Bernassola F, Ranalli M, Yee K, Zong WX, Corazzari M, et al. (2004). p73 induces apoptosis via PUMA transactivation and Bax mitochondrial translocation. J Biol Chem, 279:8076-8083.
    1. Nakano K, Vousden KH (2001). PUMA, a novel proapoptotic gene, is induced by p53. Mol Cell, 7:683-694.
    1. Fricker M, Papadia S, Hardingham GE, Tolkovsky AM (2010). Implication of TAp73 in the p53-independent pathway of Puma induction and Puma-dependent apoptosis in primary cortical neurons. J Neurochem, 114:772-783.
    1. Wilson AM, Morquette B, Abdouh M, Unsain N, Barker PA, Feinstein E, et al. (2013). ASPP1/2 regulate p53-dependent death of retinal ganglion cells through PUMA and Fas/CD95 activation in vivo. J Neurosci, 33:2205-2216.
    1. Hemann MT, Zilfou JT, Zhao Z, Burgess DJ, Hannon GJ, Lowe SW (2004). Suppression of tumorigenesis by the p53 target PUMA. Proc Natl Acad Sci U S A, 101:9333-9338.
    1. Reimertz C, Kögel D, Rami A, Chittenden T, Prehn JH (2003). Gene expression during ER stress-induced apoptosis in neurons: induction of the BH3-only protein Bbc3/PUMA and activation of the mitochondrial apoptosis pathway. J Cell Biol, 162: 587-597.
    1. Bunk EC, König HG, Bernas T, Engel T, Henshall DC, Kirby BP, et al. (2010). BH3-only proteins BIM and PUMA in the regulation of survival and neuronal differentiation of newly generated cells in the adult mouse hippocampus. Cell Death Dis, 1: e15.
    1. Liu Z, Zhang X, Huang D, Liu Y, Zhang X, Liu L, et al. (2012). Elevated expression of iASPP in head and neck squamous cell carcinoma and its clinical significance. Med Oncol, 29: 3381-3388.
    1. Morris EV, Cerundolo L, Lu M, Verrill C, Fritzsche F, White MJ, et al. (2014). Nuclear iASPP may facilitate prostate cancer progression. Cell Death Dis, 5: e1492.

Source: PubMed

3
Suscribir