Skip to main content
Top
Published in: Translational Stroke Research 6/2017

01-12-2017 | Original Article

A Combination of Three Repurposed Drugs Administered at Reperfusion as a Promising Therapy for Postischemic Brain Injury

Authors: I-Chen Yu, Ping-Chang Kuo, Jui-Hung Yen, Hallel C. Paraiso, Eric T. Curfman, Benecia C. Hong-Goka, Robert D. Sweazey, Fen-Lei Chang

Published in: Translational Stroke Research | Issue 6/2017

Login to get access

Abstract

Cerebral ischemia leads to multifaceted injury to the brain. A polytherapeutic drug that can be administered immediately after reperfusion may increase protection to the brain by simultaneously targeting multiple deleterious cascades. This study evaluated efficacy of the combination of three clinically approved drugs: lamotrigine, minocycline, and lovastatin, using two mouse models: global and focal cerebral ischemia induced by transient occlusion of the common carotid arteries or the middle cerebral artery, respectively. In vitro, the combination drug, but not single drug, protected neurons against oxygen-glucose deprivation (OGD)-induced cell death. The combination drug simultaneously targeted cell apoptosis and DNA damage induced by ischemia. Besides acting on neurons, the combination drug suppressed inflammatory processes in microglia and brain endothelial cells induced by ischemia. In a transient global ischemia model, the combination drug, but not single drug, suppressed microglial activation and inflammatory cytokine production, and reduced neuronal damage. In a transient focal ischemia model, the combination drug, but not single drug, attenuated brain infarction, suppressed infiltration of peripheral neutrophils, and reduced neurological deficits following ischemic stroke. In summary, the combination drug confers a broad-spectrum protection against ischemia/reperfusion (I/R) injury and could be a promising approach for early neuroprotection after out-of-hospital cardiac arrest or ischemic stroke.
Appendix
Available only for authorised users
Literature
2.
go back to reference Nielsen N, Wetterslev J, Cronberg T, Erlinge D, Gasche Y, Hassager C, et al. Targeted temperature management at 33 degrees C versus 36 degrees C after cardiac arrest. N Engl J Med. 2013;369(23):2197–206. doi:10.1056/NEJMoa1310519.CrossRefPubMed Nielsen N, Wetterslev J, Cronberg T, Erlinge D, Gasche Y, Hassager C, et al. Targeted temperature management at 33 degrees C versus 36 degrees C after cardiac arrest. N Engl J Med. 2013;369(23):2197–206. doi:10.​1056/​NEJMoa1310519.CrossRefPubMed
3.
go back to reference Haugk M, Testori C, Sterz F, Uranitsch M, Holzer M, Behringer W, et al. Relationship between time to target temperature and outcome in patients treated with therapeutic hypothermia after cardiac arrest. Crit Care. 2011;15(2):R101. doi:10.1186/cc10116.CrossRefPubMedPubMedCentral Haugk M, Testori C, Sterz F, Uranitsch M, Holzer M, Behringer W, et al. Relationship between time to target temperature and outcome in patients treated with therapeutic hypothermia after cardiac arrest. Crit Care. 2011;15(2):R101. doi:10.​1186/​cc10116.CrossRefPubMedPubMedCentral
7.
go back to reference Papazisis G, Kallaras K, Kaiki-Astara A, Pourzitaki C, Tzachanis D, Dagklis T, et al. Neuroprotection by lamotrigine in a rat model of neonatal hypoxic-ischaemic encephalopathy. Int J Neuropsychopharmacol. 2008;11(3):321–9. doi:10.1017/S1461145707008012.CrossRefPubMed Papazisis G, Kallaras K, Kaiki-Astara A, Pourzitaki C, Tzachanis D, Dagklis T, et al. Neuroprotection by lamotrigine in a rat model of neonatal hypoxic-ischaemic encephalopathy. Int J Neuropsychopharmacol. 2008;11(3):321–9. doi:10.​1017/​S146114570700801​2.CrossRefPubMed
8.
go back to reference Smith SE, Meldrum BS. Cerebroprotective effect of lamotrigine after focal ischemia in rats. Stroke. 1995;26(1):117–21. discussion 21-2CrossRefPubMed Smith SE, Meldrum BS. Cerebroprotective effect of lamotrigine after focal ischemia in rats. Stroke. 1995;26(1):117–21. discussion 21-2CrossRefPubMed
10.
go back to reference Ni Chroinin D, Asplund K, Asberg S, Callaly E, Cuadrado-Godia E, Diez-Tejedor E, et al. Statin therapy and outcome after ischemic stroke: systematic review and meta-analysis of observational studies and randomized trials. Stroke. 2013;44(2):448–56. doi:10.1161/STROKEAHA.112.668277.CrossRefPubMed Ni Chroinin D, Asplund K, Asberg S, Callaly E, Cuadrado-Godia E, Diez-Tejedor E, et al. Statin therapy and outcome after ischemic stroke: systematic review and meta-analysis of observational studies and randomized trials. Stroke. 2013;44(2):448–56. doi:10.​1161/​STROKEAHA.​112.​668277.CrossRefPubMed
11.
go back to reference Antoniades C, Bakogiannis C, Leeson P, Guzik TJ, Zhang MH, Tousoulis D, et al. Rapid, direct effects of statin treatment on arterial redox state and nitric oxide bioavailability in human atherosclerosis via tetrahydrobiopterin-mediated endothelial nitric oxide synthase coupling. Circulation. 2011;124(3):335–45. doi:10.1161/CIRCULATIONAHA.110.985150.CrossRefPubMedPubMedCentral Antoniades C, Bakogiannis C, Leeson P, Guzik TJ, Zhang MH, Tousoulis D, et al. Rapid, direct effects of statin treatment on arterial redox state and nitric oxide bioavailability in human atherosclerosis via tetrahydrobiopterin-mediated endothelial nitric oxide synthase coupling. Circulation. 2011;124(3):335–45. doi:10.​1161/​CIRCULATIONAHA.​110.​985150.CrossRefPubMedPubMedCentral
13.
15.
17.
go back to reference Potey C, Ouk T, Petrault O, Petrault M, Berezowski V, Salleron J, et al. Early treatment with atorvastatin exerts parenchymal and vascular protective effects in experimental cerebral ischaemia. Br J Pharmacol. 2015;172(21):5188–98. doi:10.1111/bph.13285.CrossRefPubMedPubMedCentral Potey C, Ouk T, Petrault O, Petrault M, Berezowski V, Salleron J, et al. Early treatment with atorvastatin exerts parenchymal and vascular protective effects in experimental cerebral ischaemia. Br J Pharmacol. 2015;172(21):5188–98. doi:10.​1111/​bph.​13285.CrossRefPubMedPubMedCentral
18.
go back to reference Murakami K, Kondo T, Kawase M, Chan PH. The development of a new mouse model of global ischemia: focus on the relationships between ischemia duration, anesthesia, cerebral vasculature, and neuronal injury following global ischemia in mice. Brain Res. 1998;780(2):304–10.CrossRefPubMed Murakami K, Kondo T, Kawase M, Chan PH. The development of a new mouse model of global ischemia: focus on the relationships between ischemia duration, anesthesia, cerebral vasculature, and neuronal injury following global ischemia in mice. Brain Res. 1998;780(2):304–10.CrossRefPubMed
20.
22.
go back to reference Wellons JC 3rd, Sheng H, Laskowitz DT, Mackensen GB, Pearlstein RD, Warner DS. A comparison of strain-related susceptibility in two murine recovery models of global cerebral ischemia. Brain Res. 2000;868(1):14–21.CrossRefPubMed Wellons JC 3rd, Sheng H, Laskowitz DT, Mackensen GB, Pearlstein RD, Warner DS. A comparison of strain-related susceptibility in two murine recovery models of global cerebral ischemia. Brain Res. 2000;868(1):14–21.CrossRefPubMed
23.
go back to reference Yang G, Kitagawa K, Matsushita K, Mabuchi T, Yagita Y, Yanagihara T, et al. C57BL/6 strain is most susceptible to cerebral ischemia following bilateral common carotid occlusion among seven mouse strains: selective neuronal death in the murine transient forebrain ischemia. Brain Res. 1997;752(1–2):209–18.CrossRefPubMed Yang G, Kitagawa K, Matsushita K, Mabuchi T, Yagita Y, Yanagihara T, et al. C57BL/6 strain is most susceptible to cerebral ischemia following bilateral common carotid occlusion among seven mouse strains: selective neuronal death in the murine transient forebrain ischemia. Brain Res. 1997;752(1–2):209–18.CrossRefPubMed
24.
go back to reference Longa EZ, Weinstein PR, Carlson S, Cummins R. Reversible middle cerebral artery occlusion without craniectomy in rats. Stroke. 1989;20(1):84–91.CrossRefPubMed Longa EZ, Weinstein PR, Carlson S, Cummins R. Reversible middle cerebral artery occlusion without craniectomy in rats. Stroke. 1989;20(1):84–91.CrossRefPubMed
25.
28.
go back to reference Li Y, Maher P, Schubert D. A role for 12-lipoxygenase in nerve cell death caused by glutathione depletion. Neuron. 1997;19(2):453–63.CrossRefPubMed Li Y, Maher P, Schubert D. A role for 12-lipoxygenase in nerve cell death caused by glutathione depletion. Neuron. 1997;19(2):453–63.CrossRefPubMed
29.
32.
go back to reference Goldberg MP, Choi DW. Combined oxygen and glucose deprivation in cortical cell culture: calcium-dependent and calcium-independent mechanisms of neuronal injury. J Neurosci. 1993;13(8):3510–24.PubMed Goldberg MP, Choi DW. Combined oxygen and glucose deprivation in cortical cell culture: calcium-dependent and calcium-independent mechanisms of neuronal injury. J Neurosci. 1993;13(8):3510–24.PubMed
34.
35.
go back to reference Ponomarev ED, Shriver LP, Dittel BN. CD40 expression by microglial cells is required for their completion of a two-step activation process during central nervous system autoimmune inflammation. J Immunol. 2006;176(3):1402–10.CrossRefPubMed Ponomarev ED, Shriver LP, Dittel BN. CD40 expression by microglial cells is required for their completion of a two-step activation process during central nervous system autoimmune inflammation. J Immunol. 2006;176(3):1402–10.CrossRefPubMed
37.
go back to reference Horn M, Schlote W. Delayed neuronal death and delayed neuronal recovery in the human brain following global ischemia. Acta Neuropathol. 1992;85(1):79–87.CrossRefPubMed Horn M, Schlote W. Delayed neuronal death and delayed neuronal recovery in the human brain following global ischemia. Acta Neuropathol. 1992;85(1):79–87.CrossRefPubMed
39.
go back to reference Petito CK, Feldmann E, Pulsinelli WA, Plum F. Delayed hippocampal damage in humans following cardiorespiratory arrest. Neurology. 1987;37(8):1281–6.CrossRefPubMed Petito CK, Feldmann E, Pulsinelli WA, Plum F. Delayed hippocampal damage in humans following cardiorespiratory arrest. Neurology. 1987;37(8):1281–6.CrossRefPubMed
42.
go back to reference Sutherland BA, Neuhaus AA, Couch Y, Balami JS, DeLuca GC, Hadley G, et al. The transient intraluminal filament middle cerebral artery occlusion model as a model of endovascular thrombectomy in stroke. J Cereb Blood Flow Metab. 2016;36(2):363–9. doi:10.1177/0271678X15606722.CrossRefPubMed Sutherland BA, Neuhaus AA, Couch Y, Balami JS, DeLuca GC, Hadley G, et al. The transient intraluminal filament middle cerebral artery occlusion model as a model of endovascular thrombectomy in stroke. J Cereb Blood Flow Metab. 2016;36(2):363–9. doi:10.​1177/​0271678X15606722​.CrossRefPubMed
44.
45.
go back to reference Kernt M, Hirneiss C, Neubauer AS, Kampik A. Minocycline is cytoprotective in human corneal endothelial cells and induces anti-apoptotic B-cell CLL/lymphoma 2 (Bcl-2) and X-linked inhibitor of apoptosis (XIAP). Br J Ophthalmol. 2010;94(7):940–6. doi:10.1136/bjo.2009.165092.CrossRefPubMed Kernt M, Hirneiss C, Neubauer AS, Kampik A. Minocycline is cytoprotective in human corneal endothelial cells and induces anti-apoptotic B-cell CLL/lymphoma 2 (Bcl-2) and X-linked inhibitor of apoptosis (XIAP). Br J Ophthalmol. 2010;94(7):940–6. doi:10.​1136/​bjo.​2009.​165092.CrossRefPubMed
51.
go back to reference Meske V, Albert F, Richter D, Schwarze J, Ohm TG. Blockade of HMG-CoA reductase activity causes changes in microtubule-stabilizing protein tau via suppression of geranylgeranylpyrophosphate formation: implications for Alzheimer’s disease. Eur J Neurosci. 2003;17(1):93–102.CrossRefPubMed Meske V, Albert F, Richter D, Schwarze J, Ohm TG. Blockade of HMG-CoA reductase activity causes changes in microtubule-stabilizing protein tau via suppression of geranylgeranylpyrophosphate formation: implications for Alzheimer’s disease. Eur J Neurosci. 2003;17(1):93–102.CrossRefPubMed
52.
go back to reference Russo E, di Donato Paola E, Gareri P, Siniscalchi A, Labate A, Gallelli L, et al. Pharmacodynamic potentiation of antiepileptic drugs’ effects by some HMG-CoA reductase inhibitors against audiogenic seizures in DBA/2 mice. Pharmacol Res. 2013;70(1):1–12. doi:10.1016/j.phrs.2012.12.002.CrossRefPubMed Russo E, di Donato Paola E, Gareri P, Siniscalchi A, Labate A, Gallelli L, et al. Pharmacodynamic potentiation of antiepileptic drugs’ effects by some HMG-CoA reductase inhibitors against audiogenic seizures in DBA/2 mice. Pharmacol Res. 2013;70(1):1–12. doi:10.​1016/​j.​phrs.​2012.​12.​002.CrossRefPubMed
54.
56.
go back to reference Clausen BH, Lambertsen KL, Babcock AA, Holm TH, Dagnaes-Hansen F, Finsen B. Interleukin-1beta and tumor necrosis factor-alpha are expressed by different subsets of microglia and macrophages after ischemic stroke in mice. J Neuroinflammation. 2008;5:46. doi:10.1186/1742-2094-5-46.CrossRefPubMedPubMedCentral Clausen BH, Lambertsen KL, Babcock AA, Holm TH, Dagnaes-Hansen F, Finsen B. Interleukin-1beta and tumor necrosis factor-alpha are expressed by different subsets of microglia and macrophages after ischemic stroke in mice. J Neuroinflammation. 2008;5:46. doi:10.​1186/​1742-2094-5-46.CrossRefPubMedPubMedCentral
61.
63.
go back to reference Maynard C, Longstreth WT Jr, Nichol G, Hallstrom A, Kudenchuk PJ, Rea T, et al. Effect of prehospital induction of mild hypothermia on 3-month neurological status and 1-year survival among adults with cardiac arrest: long-term follow-up of a randomized, clinical trial. J Am Heart Assoc. 2015;4(3):e001693. doi:10.1161/JAHA.114.001693.CrossRefPubMedPubMedCentral Maynard C, Longstreth WT Jr, Nichol G, Hallstrom A, Kudenchuk PJ, Rea T, et al. Effect of prehospital induction of mild hypothermia on 3-month neurological status and 1-year survival among adults with cardiac arrest: long-term follow-up of a randomized, clinical trial. J Am Heart Assoc. 2015;4(3):e001693. doi:10.​1161/​JAHA.​114.​001693.CrossRefPubMedPubMedCentral
68.
go back to reference Ebinger M, Winter B, Wendt M, Weber JE, Waldschmidt C, Rozanski M, et al. Effect of the use of ambulance-based thrombolysis on time to thrombolysis in acute ischemic stroke: a randomized clinical trial. JAMA. 2014;311(16):1622–31. doi:10.1001/jama.2014.2850.CrossRefPubMed Ebinger M, Winter B, Wendt M, Weber JE, Waldschmidt C, Rozanski M, et al. Effect of the use of ambulance-based thrombolysis on time to thrombolysis in acute ischemic stroke: a randomized clinical trial. JAMA. 2014;311(16):1622–31. doi:10.​1001/​jama.​2014.​2850.CrossRefPubMed
Metadata
Title
A Combination of Three Repurposed Drugs Administered at Reperfusion as a Promising Therapy for Postischemic Brain Injury
Authors
I-Chen Yu
Ping-Chang Kuo
Jui-Hung Yen
Hallel C. Paraiso
Eric T. Curfman
Benecia C. Hong-Goka
Robert D. Sweazey
Fen-Lei Chang
Publication date
01-12-2017
Publisher
Springer US
Published in
Translational Stroke Research / Issue 6/2017
Print ISSN: 1868-4483
Electronic ISSN: 1868-601X
DOI
https://doi.org/10.1007/s12975-017-0543-5

Other articles of this Issue 6/2017

Translational Stroke Research 6/2017 Go to the issue