Hibernation-like state induced by an opioid peptide protects against experimental stroke

Cesar V Borlongan, Teruo Hayashi, Peter R Oeltgen, Tsung-Ping Su, Yun Wang, Cesar V Borlongan, Teruo Hayashi, Peter R Oeltgen, Tsung-Ping Su, Yun Wang

Abstract

Background: Delta opioid peptide [D-ala2,D-leU5]enkephalin (DADLE) induces hibernation in summer ground squirrels, and enhances preservation and survival of isolated or transplanted lungs and hearts. In the present study, we investigated the protective effect of DADLE in the central nervous system.

Results: Adult Sprague-Dawley rats were pretreated with DADLE (4 mg/kg every 2 h x 4 injections, i.p.) or saline prior to unilateral occlusion of the middle cerebral artery (MCA). Daily behavioral tests revealed that ischemic animals treated with DADLE did not show any significant behavioral dysfunctions compared with saline-treated ischemic animals. Opioid antagonists only transiently inhibited the protective effect of DADLE, indicating the participation of non-opioid mechanisms in DADLE neuroprotection. Histological examination using triphenyltetrazolium chloride (TTC) revealed that brains from ischemic animals treated with DADLE, either alone or with adjuvant opioid blockers, exhibited almost completely intact striata. In contrast, brains from ischemic animals that received saline showed significant infarction in the lateral striatum. Analyses of apoptotic cell death revealed a significant increase in the p-53 mRNA expression in the striatum of ischemic animals that received saline, while those that received DADLE exhibited near normal striatal p-53 expression. This protective effect was accompanied by significant increments in protein levels of glial cell line-derived neurotrophic factor in the striatum of DADLE-treated ischemic animals.

Conclusion: These results indicate that DADLE protected against necrotic and apoptotic cell death processes associated with ischemia-reperfusion injury. The present study demonstrates that delta opioids are crucially involved in stroke, suggesting that the opioid system is important in the study of brain injury and protection.

Figures

Figure 1
Figure 1
[D-ala2,D-leU5]enkephalin (DADLE) attenuates ischemia-induced asymmetrical motor deficits. Ischemic animals pretreated with saline (S), naltrexone alone (N) or naloxone methiodide alone (NM) displayed significant biased swing activity (panel A), postural bias (panel B), spontaneous rotational behavior (panel C), and forelimb akinesia (panel D). In contrast, ischemic animals pretreated with DADLE + naltrexone (D + N) or DADLE + naloxone methiodide (D + NM) exhibited significant dysfunctions in the same behavioral tests only at 0 to 24 h post occlusion and reperfusion surgery of the middle cerebral artery (MCAor) (days 0 and 1), and thereafter showed near normal behaviors at 48 h and 72 h post MCAor (days 2 and 3). In contrast, ischemic animals pretreated with DADLE alone (D) displayed near normal behaviors throughout the post-MCAor test period.
Figure 2
Figure 2
[D-ala2,D-leU5]enkephalin (DADLE) reduces necrotic and apoptotic (panel B) cell death associated with ischemia. (A) Triphenyltetrazolium chloride (TTC) staining at 72 h post occlusion and reperfusion surgery of the middle cerebral artery (MCAor) revealed that the striata from ischemic animals exposed to ischemia/reperfusion surgery and pretreated with saline showed dehydrogenase-deficient tissue (negative TTC stains). In contrast, the striata from ischemic animals pretreated with DADLE, DADLE + naloxone methiodide, or DADLE + naltrexone did not reveal any dehydrogenase deficiency. (B) Ischemic animals exposed to ischemia/reperfusion injury and treated with saline showed a significant increase in mRNA expression of p-53 in their ischemic striata (65% increment compared with intact striatum of normal, control animals) at 24 h after stroke surgery. In contrast, ischemic animals treated with DADLE exhibited only a small increment in p-53 mRNA expression in their ischemic striata at the same time period (28% increment compared with intact striatum of normal, control animals; not significantly different from control values). Comparisons of ischemic striata between these two groups showed a marked reduction in p-53 mRNA expression in DADLE-treated ischemic animals compared with saline-treated ischemic animals. (C) Immunohistochemical analyses of phenotypic markers of apoptosis revealed that DADLE significantly reduced caspase-3- (panels a, b, c, d) and Fas-positive cells (panels e, f, g, h) in DADLE-treated ischemic animals compared to saline-treated ischemic animals. Quantitative data shown in bar graphs and represent means ± S.E.M. Asterisks correspond to statistical significance at P <0.05. To better capture the necrotic and apoptotic cells in saline-treated ischemic animals (panel d), higher magnification images are generated from propidium iodide (a) and caspase-3 (b) immunofluorescently labeled striatal cells, respectively. Scale bar = 50 μm (a), 100 μm (b).
Figure 3
Figure 3
[D-ala2,D-leU5]enkephalin (DADLE) increases expression of glial cell line-derived neurotrophic factor (GDNF) in brain tissues. Enzyme-linked immunosorbent assays (ELISA) revealed that the levels of GDNF proteins, but not nerve growth factor (NGF), were significantly higher in striatal and cortical tissues harvested from ischemic animals treated with DADLE compared with those treated with saline. Data are expressed as mean percent of control ± S.E.M. Asterisk indicates P <0.05. n = 6 samples for each neurotrophic factor examined.
Figure 4
Figure 4
Schematic diagram summarizing timeline of experimental procedures in this study.

References

    1. Maldonado R, Saiardi A, Valverde O, Samad TA, Roques BP, Borrelli E. Absence of opiate rewarding effects in mice lacking dopamine D2 receptors. Nature. 1997;388:586–589. doi: 10.1038/41567.
    1. Cook CD, Rodefer JS, Picker MJ. Selective attenuation of the antinociceptive effects of mu opioids by the putative dopamine D3 agonist 7-OH-DPAT. Psychopharmacology. 1999;144:239–247. doi: 10.1007/s002130050999.
    1. Boutin H, Dauphin F, MacKenzie ET, Jauzac P. Differential time-course decreases in nonselective, mu-, delta-, and kappa-opioid receptors after focal cerebral ischemia in mice. Stroke. 1999;30:1271–1277.
    1. Frerichs KU, Hallenbeck JM. Hibernation in ground squirrels induces state and species-specific tolerance to hypoxia and aglycemia: an in vitro study in hippocampal slices. J Cereb Blood Flow Metab. 1998;18:168–175. doi: 10.1097/00004647-199802000-00007.
    1. Kevelaitis E, Peynet J, Mouas C, Launay JM, Menasche P. Opening of potassium channels: the common cardioprotective link between preconditioning and natural hibernation? Circulation. 1999;99:3079–3085.
    1. Wang Y, Lin SZ, Chiou AL, Williams LR, Hoffer BJ. Glial cell line-derived neurotrophic factor protects against ischemia-induced injury in the cerebral cortex. J Neurosci. 1997;17:4341–4348.
    1. Hayashi T, Sakai K, Sasaki C, Itoyama Y, Abe K. Loss of bag-1 immunoreactivity in rat brain after transient middle cerebral artery occlusion. Brain Res. 2000;852:496–500. doi: 10.1016/S0006-8993(99)02266-0.
    1. Wang Y, Chang CF, Morales M, Chou J, Chen HL, Chiang YH, Lin SZ, Cadet JL, Deng X, Wang JY, Chen SY, Kaplan PL, Hoffer BJ. Bone morphogenetic protein-6 reduces ischemia-induced brain damage in rats. Stroke. 2001;32:2170–2178. doi: 10.1161/hs0901.095650.
    1. Yin D, Mufson RA, Wang R, Shi Y. Fas-mediated cell death promoted by opioids. Nature. 1999;397:218. doi: 10.1038/16612.
    1. Shibata S, Tominaga K, Watanabe S. kappa-Opioid receptor agonist protects against ischemic reduction of 2-deoxyglucose uptake in morphine-tolerant rats. Eur J Pharmacol. 1995;279:197–202. doi: 10.1016/0014-2999(95)00152-B.
    1. Woodburn VL, Oles R, Poat J, Woodruff GN, Hughes J. Anticonvulsant agents, dizocilpine maleate, enadoline and HA 966 have different effects on N-methyl-DL-aspartate-induced immediate early gene induction in mice. Neuroscience. 1993;56:703–709. doi: 10.1016/0306-4522(93)90367-O.
    1. Hayashi T, Tsao LI, Cadet JL, Su TP. D-Ala2, D-Leu5]enkephalin blocks the methamphetamine-induced c-fos mRNA increase in mouse striatum. Eur J Pharmacol. 1999;366:7–8. doi: 10.1016/S0014-2999(98)00947-9.
    1. Tsao L, Cadet JL, Su TP. Reversal by [D-Ala2,D-Leu5]enkephalin of the dopamine transporter loss caused by methamphetamine. Eur J Pharmacol. 1999;372:5–7. doi: 10.1016/S0014-2999(99)00240-X.
    1. Tsao L, Ladenheim B, Andrews AM, Chiueh CC, Cadet JL, Su TP. Delta opioid peptide [D-Ala2,D-leu5]enkephalin blocks the long-term loss of dopamine transporters induced by multiple administrations of methamphetamine: involvement of opioid receptors and reactive oxygen species. J Pharmacol Exp Ther. 1998;287:322–331.
    1. Butterfield DA, Koppal T, Howard B, Subramaniam R, Hall N, Hensley K, Yatin S, Allen K, Aksenov M, Aksenova M, Carney J. Structural and functional changes in proteins induced by free radical-mediated oxidative stress and protective action of the antioxidants N-tert-butyl-alpha-phenylnitrone and vitamin E. Ann NY Acad Sci. 1998;854:448–462. doi: 10.1111/j.1749-6632.1998.tb09924.x.
    1. Fujimura M, Morita-Fujimura Y, Kawase M, Copin JC, Calagui B, Epstein CJ, Chan PH. Manganese superoxide dismutase mediates the early release of mitochondrial cytochrome C and subsequent DNA fragmentation after permanent focal cerebral ischemia in mice. J Neurosci. 1999;19:3414–3422.
    1. Borlongan CV, Tajima Y, Trojanowski JQ, Lee VM, Sanberg PR. Transplantation of cryopreserved human embryonal carcinoma-derived neurons (NT2N cells) promotes functional recovery in ischemic rats. Exp Neurol. 1998;149:310–321. doi: 10.1006/exnr.1997.6730.
    1. Onizuka K, Fukuda A, Kunimatsu M, Kumazaki M, Sasaki M, Takaku A, Nishino H. Early cytopathic features in rat ischemia model and reconstruction by neural graft. Exp Neurol. 1996;137:324–332. doi: 10.1006/exnr.1996.0033.
    1. Nishino H, Czurko A, Onizuka K, Fukuda A, Hida H, Ungsuparkorn C, Kunimatsu M, Sasaki M, Karádi Z, Lénárd L. Neuronal damage following transient cerebral ischemia and its restoration by neural transplant. Neurobiology. 1994;2:223–234.
    1. Borlongan CV, Zhou FC, Hayashi T, Su TP, Hoffer BJ, Wang Y. Involvement of GDNF in neuronal protection against 6-OHDA-induced parkinsonism following intracerebral transplantation of fetal kidney tissues in adult rats. Neurobiol Dis. 2001;8:636–646. doi: 10.1006/nbdi.2001.0410.
    1. Lin LF, Doherty DH, Lile JD, Bektesh S, Collins F. GDNF: a glial cell line-derived neurotrophic factor for midbrain dopaminergic neurons. Science. 1993;260:1130–1132. doi: 10.1126/science.8493557.
    1. Borlongan CV, Su TP, Wang Y. Treatment with delta opioid peptide enhances in vitro and in vivo survival of rat dopaminergic neurons. Neuroreport. 2000;11:923–926.
    1. Borlongan CV, Su TP, Wang Y. Delta opioid peptide augments functional effects and intrastriatal graft survival of rat fetal ventral mesencephalic cells. Cell Transplant. 2001;10:53–58.
    1. Paratcha G, Ibáñez CF, Ledda F. GDNF is a chemoattractant factor for neuronal precursor cells in the rostral migratory stream. Mol Cell Neurosci. 2006;31:505–514. doi: 10.1016/j.mcn.2005.11.007.
    1. Kobayashi T, Ahlenius H, Thored P, Kobayashi R, Kokaia Z, Lindvall O. Intracerebral infusion of glial cell line-derived neurotrophic factor promotes striatal neurogenesis after stroke in adult rats. Stroke. 2006;37:2361–2367. doi: 10.1161/01.STR.0000236025.44089.e1.
    1. Stenman E, Jamali R, Henriksson M, Maddahi A, Edvinsson L. Cooperative effect of angiotensin AT(1) and endothelin ET(A) receptor antagonism limits the brain damage after ischemic stroke in rat. Eur J Pharmacol. 2007;570:142–148. doi: 10.1016/j.ejphar.2007.05.049.
    1. Hossmann KA. Pathophysiology and therapy of experimental stroke. Cell Mol Neurobiol. 2006;26:1057–1083. doi: 10.1007/s10571-006-9008-1.
    1. Kearns CM, Cass WA, Smoot K, Kryscio R, Gash DM. GDNF protection against 6-OHDA: time dependence and requirement for protein synthesis. J Neurosci. 1997;17:7111–7118.
    1. Fox CM, Gash DM, Smoot MK, Cass WA. Neuroprotective effects of GDNF against 6-OHDA in young and aged rats. Brain Res. 2001;896:56–63. doi: 10.1016/S0006-8993(00)03270-4.
    1. Frost JJ, Wagner HN, Jr, Dannals RF, Ravert HT, Links JM, Wilson AA, Burns HD, Wong DF, McPherson RW, Rosenbaum AE. Imaging opiate receptors in the human brain by positron tomography. J Comput Assist Tomogr. 1985;9:231–236. doi: 10.1097/00004728-198503000-00001.
    1. Shacoori V, Saiag B, Rault B. Melatonin modifies prolactin release induced by opiate antagonists in male rats. Endocr Res. 1995;21:545–553.
    1. Nikkhah G, Cunningham MG, McKay R, Bjorklund A. Dopaminergic microtransplants into the substantia nigra of neonatal rats with bilateral 6-OHDA lesions. II, Transplant-induced behavioral recovery. J Neurosci. 1995;15:3562–3570.
    1. Bederson JB, Pitts LH, Tsuji M, Nishimura MC, Davis RL, Bartkowiski HM. Rat middle cerebral artery occlusion: evaluation of the model and development of a neurologic examination. Stroke. 1986;17:472–476.
    1. Bienz B, Zakut-Houri R, Givol D, Oren M. Analysis of the gene coding for the murine cellular tumour antigen p53. EMBO J. 1984;3:2179–2183.
    1. Chomczynski P, Sacchi N. Single-step method of RNA isolation by acid guanidinium thiocyanate-phenol-chloroform extraction. Anal Biochem. 1987;162:156–159. doi: 10.1016/0003-2697(87)90021-2.
    1. Mattson MP. Neuroprotective signaling and the aging brain: take away my food and let me run. Brain Res. 2000;886:47–53. doi: 10.1016/S0006-8993(00)02790-6.
    1. Okragly AJ, Haak-Frendscho M. An acid-treatment method for the enhanced detection of GDNF in biological samples. Exp Neurol. 1997;145:592–596. doi: 10.1006/exnr.1997.6500.

Source: PubMed

3
Subskrybuj