Skip to main content
Top
Published in: Translational Stroke Research 1/2016

Open Access 01-02-2016 | Original Article

Co-ultramicronized Palmitoylethanolamide/Luteolin in the Treatment of Cerebral Ischemia: from Rodent to Man

Authors: Carlo Caltagirone, Carlo Cisari, Carlo Schievano, Rosanna Di Paola, Marika Cordaro, Giuseppe Bruschetta, Emanuela Esposito, Salvatore Cuzzocrea, Stroke Study Group

Published in: Translational Stroke Research | Issue 1/2016

Login to get access

Abstract

Acute ischemic stroke, the most frequent cause of permanent disability in adults worldwide, results from transient or permanent reduction in regional cerebral blood flow and involves oxidative stress and inflammation. Despite the success of experimental animal models of stroke in identifying anti-inflammatory/neuroprotective compounds, translation of these putative neuroprotectants to human clinical trials has failed to produce a positive outcome. Tissue injury and stress activate endogenous mechanisms which function to restore homeostatic balance and prevent further damage by upregulating the synthesis of lipid signaling molecules, including N-palmitoylethanolamine (PEA or palmitoylethanolamide). PEA exerts neuroprotection and reduces inflammatory secondary events associated with brain ischemia reperfusion injury (middle cerebral artery occlusion (MCAo)). Here, we examined the neuroprotective potential of a co-ultramicronized composite containing PEA and the antioxidant flavonoid luteolin (10:1 by mass), nominated co-ultraPEALut. The study consisted of two arms. In the first, rats subjected to MCAo and treated with co-ultraPEALut post-ischemia showed reduced edema and brain infract volume, improved neurobehavioral functions, and reduced expression of pro-inflammatory markers and astrocyte markers. In the second arm, a cohort of 250 stroke patients undergoing neurorehabilitation on either an inpatient or outpatient basis were treated for 60 days with a pharmaceutical preparation of co-ultraPEALut (Glialia®). At baseline and after 30 days of treatment, all patients underwent a battery of evaluations to assess neurological status, impairment of cognitive abilities, the degree of spasticity, pain, and independence in daily living activities. All indices showed statistically significant gains at study end. Despite its observational nature, this represents the first description of co-ultraPEALut administration to human stroke patients and clinical improvement not otherwise expected from spontaneous recovery. Further, controlled trials are warranted to confirm the utility of co-ultraPEALut to improve clinical outcome in human stroke.
Literature
1.
go back to reference Thrift AG, Cadilhac DA, Thayabaranathan T, Howard G, Howard VJ, Rothwell PM, et al. Global stroke statistics. Int J Stroke. 2014;9:6–18.CrossRefPubMed Thrift AG, Cadilhac DA, Thayabaranathan T, Howard G, Howard VJ, Rothwell PM, et al. Global stroke statistics. Int J Stroke. 2014;9:6–18.CrossRefPubMed
2.
go back to reference American Heart Association Statistics Committee and Stroke Statistics Committee. Heart disease and stroke statistics—2014 update: a report from the American Heart Association. Circulation. 2014;129(3):e28–292.CrossRef American Heart Association Statistics Committee and Stroke Statistics Committee. Heart disease and stroke statistics—2014 update: a report from the American Heart Association. Circulation. 2014;129(3):e28–292.CrossRef
3.
go back to reference Corbett D, Jeffers M, Nguemeni C, Gomez-Smith M, Livingston-Thomas J. Lost in translation: rethinking approaches to stroke recovery. Prog Brain Res. 2015;218:413–34.CrossRefPubMed Corbett D, Jeffers M, Nguemeni C, Gomez-Smith M, Livingston-Thomas J. Lost in translation: rethinking approaches to stroke recovery. Prog Brain Res. 2015;218:413–34.CrossRefPubMed
5.
6.
go back to reference Chew LJ, Takanohashi A, Bell M. Microglia and inflammation: impact on developmental brain injuries. Ment Retard Dev Disabil Res Rev. 2006;12:105–12.CrossRefPubMed Chew LJ, Takanohashi A, Bell M. Microglia and inflammation: impact on developmental brain injuries. Ment Retard Dev Disabil Res Rev. 2006;12:105–12.CrossRefPubMed
7.
go back to reference Hanisch U-K, Kettenmann H. Microglia: active sensor and versatile effector cells in the normal and pathologic brain. Nat Neurosci. 2007;10:1387–94.CrossRefPubMed Hanisch U-K, Kettenmann H. Microglia: active sensor and versatile effector cells in the normal and pathologic brain. Nat Neurosci. 2007;10:1387–94.CrossRefPubMed
8.
go back to reference Jin Y, Silverman AJ, Vannucci SJ. Mast cells are early responders after hypoxia-ischemia in immature rat brain. Stroke. 2009;40:3107–12.CrossRefPubMed Jin Y, Silverman AJ, Vannucci SJ. Mast cells are early responders after hypoxia-ischemia in immature rat brain. Stroke. 2009;40:3107–12.CrossRefPubMed
9.
go back to reference Silver R, Curley JP. Mast cells on the mind: new insights and opportunities. Trends Neurosci. 2013;36:513–21.CrossRefPubMed Silver R, Curley JP. Mast cells on the mind: new insights and opportunities. Trends Neurosci. 2013;36:513–21.CrossRefPubMed
11.
go back to reference Calabresi P, Cupini LM, Centonze D, Pisani F, Bernardi G. Antiepileptic drugs as a possible neuroprotective strategy in brain ischemia. Ann Neurol. 2003;5:693–702.CrossRef Calabresi P, Cupini LM, Centonze D, Pisani F, Bernardi G. Antiepileptic drugs as a possible neuroprotective strategy in brain ischemia. Ann Neurol. 2003;5:693–702.CrossRef
12.
go back to reference ArunaDevi R, Lata S, Bhadoria BK, Ramteke VD, Kumar S, Sankar P, et al. Neuroprotective effect of 5,7,3′,4′,5′-pentahydroxy dihydroflavanol-3-O-(2″-O-galloyl)-beta-D-glucopyranoside, a polyphenolic compound in focal cerebral ischemia in rat. Eur J Pharmacol. 2010;626:205–12.CrossRefPubMed ArunaDevi R, Lata S, Bhadoria BK, Ramteke VD, Kumar S, Sankar P, et al. Neuroprotective effect of 5,7,3′,4′,5′-pentahydroxy dihydroflavanol-3-O-(2″-O-galloyl)-beta-D-glucopyranoside, a polyphenolic compound in focal cerebral ischemia in rat. Eur J Pharmacol. 2010;626:205–12.CrossRefPubMed
13.
go back to reference ArunaDevi R, Ramteke VD, Kumar S, Shukla MK, Jaganathan S, Kumar D, et al. Neuroprotective effect of s-methylisothiourea in transient focal cerebral ischemia in rat. Nitric Oxide. 2010;22:1–10.CrossRefPubMed ArunaDevi R, Ramteke VD, Kumar S, Shukla MK, Jaganathan S, Kumar D, et al. Neuroprotective effect of s-methylisothiourea in transient focal cerebral ischemia in rat. Nitric Oxide. 2010;22:1–10.CrossRefPubMed
14.
go back to reference Ye Q, Li Q, Zhou Y, Xu L, Mao W, Gao Y, et al. Synthesis and evaluation of 3-(furo[2,3-b]pyridin-3-yl)-4-(1H-indol-3-yl)-maleimides as novel GSK-3β inhibitors and anti-ischemic agents. Chem Biol Drug Des. 2015. doi:10.1111/cbdd.12546. Ye Q, Li Q, Zhou Y, Xu L, Mao W, Gao Y, et al. Synthesis and evaluation of 3-(furo[2,3-b]pyridin-3-yl)-4-(1H-indol-3-yl)-maleimides as novel GSK-3β inhibitors and anti-ischemic agents. Chem Biol Drug Des. 2015. doi:10.​1111/​cbdd.​12546.
16.
go back to reference Sughrue ME, Grobelny BT, Ducruet AF, Komotar RJ, Mocco J, Sciacca RR, et al. Data presentation in rodent stroke studies and the predictive value of confidence intervals. J Clin Neurosci. 2010;17:11–5.CrossRefPubMed Sughrue ME, Grobelny BT, Ducruet AF, Komotar RJ, Mocco J, Sciacca RR, et al. Data presentation in rodent stroke studies and the predictive value of confidence intervals. J Clin Neurosci. 2010;17:11–5.CrossRefPubMed
17.
go back to reference Hussain MS, Shuaib A. Research into neuroprotection must continue … but with a different approach. Stroke. 2008;39:521–2.CrossRefPubMed Hussain MS, Shuaib A. Research into neuroprotection must continue … but with a different approach. Stroke. 2008;39:521–2.CrossRefPubMed
18.
go back to reference Prestigiacomo CJ, Kim SC, Connolly Jr ES, Liao H, Yan SF, Pinsky DJ. CD18-mediated neutrophil recruitment contributes to the pathogenesis of reperfused but not nonreperfused stroke. Stroke. 1999;30:1110–7.CrossRefPubMed Prestigiacomo CJ, Kim SC, Connolly Jr ES, Liao H, Yan SF, Pinsky DJ. CD18-mediated neutrophil recruitment contributes to the pathogenesis of reperfused but not nonreperfused stroke. Stroke. 1999;30:1110–7.CrossRefPubMed
19.
go back to reference Zhang L, Zhang ZG, Zhang RL, Lu M, Krams M, Chopp M. Effects of a selective CD11b/CD18 antagonist and recombinant human tissue plasminogen activator treatment alone and in combination in a rat embolic model of stroke. Stroke. 2003;34:1790–5.CrossRefPubMed Zhang L, Zhang ZG, Zhang RL, Lu M, Krams M, Chopp M. Effects of a selective CD11b/CD18 antagonist and recombinant human tissue plasminogen activator treatment alone and in combination in a rat embolic model of stroke. Stroke. 2003;34:1790–5.CrossRefPubMed
20.
go back to reference Enlimomab Acute Stroke Trial Investigators. Use of anti-ICAM-1 therapy in ischemic stroke: results of the Enlimomab Acute Stroke Trial. Neurology. 2001;57:1428–34.CrossRef Enlimomab Acute Stroke Trial Investigators. Use of anti-ICAM-1 therapy in ischemic stroke: results of the Enlimomab Acute Stroke Trial. Neurology. 2001;57:1428–34.CrossRef
21.
go back to reference Becker KJ. Anti-leukocyte antibodies: LeukArrest (Hu23F2G) and enlimomab (R6.5) in acute stroke. Curr Med Res Opin. 2002;18 Suppl 2:S18–22.CrossRefPubMed Becker KJ. Anti-leukocyte antibodies: LeukArrest (Hu23F2G) and enlimomab (R6.5) in acute stroke. Curr Med Res Opin. 2002;18 Suppl 2:S18–22.CrossRefPubMed
22.
go back to reference Buckley CD, Gilroy DW, Serhan CN, Stockinger B, Tak PP. The resolution of inflammation. Nat Rev Immunol. 2013;13:59–66.CrossRefPubMed Buckley CD, Gilroy DW, Serhan CN, Stockinger B, Tak PP. The resolution of inflammation. Nat Rev Immunol. 2013;13:59–66.CrossRefPubMed
24.
go back to reference Lambert DM, Vandevoorde S, Jonsson KO, Fowler CJ. The palmitoylethanolamide family: a new class of anti-inflammatory agents? Curr Med Chem. 2002;9:663–74.CrossRefPubMed Lambert DM, Vandevoorde S, Jonsson KO, Fowler CJ. The palmitoylethanolamide family: a new class of anti-inflammatory agents? Curr Med Chem. 2002;9:663–74.CrossRefPubMed
25.
go back to reference Skaper SD, Facci L. Mast cell-glia axis in neuroinflammation and therapeutic potential of the anandamide congener palmitoylethanolamide. Philos Trans R Soc Lond B Biol Sci. 2012;367:3312–25.PubMedCentralCrossRefPubMed Skaper SD, Facci L. Mast cell-glia axis in neuroinflammation and therapeutic potential of the anandamide congener palmitoylethanolamide. Philos Trans R Soc Lond B Biol Sci. 2012;367:3312–25.PubMedCentralCrossRefPubMed
27.
go back to reference Impellizzeri D, Esposito E, Attley J, Cuzzocrea S. Targeting inflammation: new therapeutic approaches in chronic kidney disease (CKD). Pharmacol Res. 2014;81:91–102.CrossRefPubMed Impellizzeri D, Esposito E, Attley J, Cuzzocrea S. Targeting inflammation: new therapeutic approaches in chronic kidney disease (CKD). Pharmacol Res. 2014;81:91–102.CrossRefPubMed
28.
go back to reference Alhouayek M, Muccioli GG. Harnessing the anti-inflammatory potential of palmitoylethanolamide. Drug Discov Today. 2014;19:1632–9.CrossRefPubMed Alhouayek M, Muccioli GG. Harnessing the anti-inflammatory potential of palmitoylethanolamide. Drug Discov Today. 2014;19:1632–9.CrossRefPubMed
29.
go back to reference Esposito E, Cordaro M, Cuzzocrea S. Roles of fatty acid ethanolamides (FAE) in traumatic and ischemic brain injury. Pharmacol Res. 2014;86:26–31.CrossRefPubMed Esposito E, Cordaro M, Cuzzocrea S. Roles of fatty acid ethanolamides (FAE) in traumatic and ischemic brain injury. Pharmacol Res. 2014;86:26–31.CrossRefPubMed
30.
go back to reference Fidaleo M, Fanelli F, Ceru MP, Moreno S. Neuroprotective properties of peroxisome proliferator-activated receptor alpha (PPARα) and its lipid ligands. Curr Med Chem. 2014;21:2803–21.CrossRefPubMed Fidaleo M, Fanelli F, Ceru MP, Moreno S. Neuroprotective properties of peroxisome proliferator-activated receptor alpha (PPARα) and its lipid ligands. Curr Med Chem. 2014;21:2803–21.CrossRefPubMed
31.
go back to reference Crupi R, Paterniti I, Ahmad A, Campolo M, Esposito E, Cuzzocrea S. Effects of palmitoylethanolamide and luteolin in an animal model of anxiety/depression. CNS Neurol Disord: Drug Targets. 2013;12:989–1001.CrossRef Crupi R, Paterniti I, Ahmad A, Campolo M, Esposito E, Cuzzocrea S. Effects of palmitoylethanolamide and luteolin in an animal model of anxiety/depression. CNS Neurol Disord: Drug Targets. 2013;12:989–1001.CrossRef
32.
go back to reference Paterniti I, Cordaro M, Campolo M, Siracusa R, Cornelius C, Navarra M, et al. Neuroprotection by association of palmitoylethanolamide with luteolin in experimental Alzheimer’s disease models: the control of neuroinflammation. CNS Neurol Disord: Drug Targets. 2014;13:1530–41.CrossRef Paterniti I, Cordaro M, Campolo M, Siracusa R, Cornelius C, Navarra M, et al. Neuroprotection by association of palmitoylethanolamide with luteolin in experimental Alzheimer’s disease models: the control of neuroinflammation. CNS Neurol Disord: Drug Targets. 2014;13:1530–41.CrossRef
33.
34.
go back to reference Cordaro M, Impellizzeri D, Paterniti I, Bruschetta G, Siracusa R, De Stefano D, et al. Neuroprotective effects of Co-ultraPEALut on secondary inflammatory process and autophagy involved in traumatic brain injury. J Neurotrauma. 2015. doi:10.1089/neu.2014.3460.PubMed Cordaro M, Impellizzeri D, Paterniti I, Bruschetta G, Siracusa R, De Stefano D, et al. Neuroprotective effects of Co-ultraPEALut on secondary inflammatory process and autophagy involved in traumatic brain injury. J Neurotrauma. 2015. doi:10.​1089/​neu.​2014.​3460.PubMed
35.
go back to reference Longa EZ, Weinstein PR, Carlson S, Cummins R. Reversible middle cerebral artery occlusion without craniectomy in rats. Stroke. 1989;20:84–91.CrossRefPubMed Longa EZ, Weinstein PR, Carlson S, Cummins R. Reversible middle cerebral artery occlusion without craniectomy in rats. Stroke. 1989;20:84–91.CrossRefPubMed
36.
go back to reference Melani A, Pantoni L, Corsi C, Bianchi L, Monopoli A, Bertorelli R, et al. Striatal outflow of adenosine, excitatory amino acids, gamma-aminobutyric acid, and taurine in awake freely moving rats after middle cerebral artery occlusion: correlations with neurological deficit and histopathological damage. Stroke. 1999;30:2448–54.CrossRefPubMed Melani A, Pantoni L, Corsi C, Bianchi L, Monopoli A, Bertorelli R, et al. Striatal outflow of adenosine, excitatory amino acids, gamma-aminobutyric acid, and taurine in awake freely moving rats after middle cerebral artery occlusion: correlations with neurological deficit and histopathological damage. Stroke. 1999;30:2448–54.CrossRefPubMed
37.
go back to reference Guo C, Yin Y, Duan J, Zhu Y, Yan J, Wei G, et al. Neuroprotective effect and underlying mechanism of sodium danshensu [3-(3,4-dihydroxyphenyl) lactic acid from Radix and Rhizoma Salviae miltiorrhizae = Danshen] against cerebral ischemia and reperfusion injury in rats. Phytomedicine. 2015;22:283–9.CrossRefPubMed Guo C, Yin Y, Duan J, Zhu Y, Yan J, Wei G, et al. Neuroprotective effect and underlying mechanism of sodium danshensu [3-(3,4-dihydroxyphenyl) lactic acid from Radix and Rhizoma Salviae miltiorrhizae = Danshen] against cerebral ischemia and reperfusion injury in rats. Phytomedicine. 2015;22:283–9.CrossRefPubMed
38.
go back to reference Lu XC, Massuda E, Lin Q, Li W, Li JH, Zhang J. Post-treatment with a novel PARG inhibitor reduces infarct in cerebral ischemia in the rat. Brain Res. 2003;978:99–103.CrossRefPubMed Lu XC, Massuda E, Lin Q, Li W, Li JH, Zhang J. Post-treatment with a novel PARG inhibitor reduces infarct in cerebral ischemia in the rat. Brain Res. 2003;978:99–103.CrossRefPubMed
39.
go back to reference Impellizzeri D, Bruschetta G, Cordaro M, Crupi R, Siracusa R, Esposito E, et al. Micronized/ultramicronized palmitoylethanolamide displays superior oral efficacy compared to nonmicronized palmitoylethanolamide in a rat model of inflammatory pain. J Neuroinflammation. 2014;11:136. doi:10.1186/s12974-014-0136-0.PubMedCentralCrossRefPubMed Impellizzeri D, Bruschetta G, Cordaro M, Crupi R, Siracusa R, Esposito E, et al. Micronized/ultramicronized palmitoylethanolamide displays superior oral efficacy compared to nonmicronized palmitoylethanolamide in a rat model of inflammatory pain. J Neuroinflammation. 2014;11:136. doi:10.​1186/​s12974-014-0136-0.PubMedCentralCrossRefPubMed
40.
go back to reference Melani A, Pantoni L, Bordoni F, Gianfriddo M, Bianchi L, Vannucchi MG, et al. The selective A2A receptor antagonist SCH 58261 reduces striatal transmitter outflow, turning behavior and ischemic brain damage induced by permanent focal ischemia in the rat. Brain Res. 2003;959:243–50.CrossRefPubMed Melani A, Pantoni L, Bordoni F, Gianfriddo M, Bianchi L, Vannucchi MG, et al. The selective A2A receptor antagonist SCH 58261 reduces striatal transmitter outflow, turning behavior and ischemic brain damage induced by permanent focal ischemia in the rat. Brain Res. 2003;959:243–50.CrossRefPubMed
41.
go back to reference Garcia JH, Wagner S, Liu KF, Hu XJ. Neurological deficit and extent of neuronal necrosis attributable to middle cerebral artery occlusion in rats. Statistical validation. Stroke. 1995;26:627–34.CrossRefPubMed Garcia JH, Wagner S, Liu KF, Hu XJ. Neurological deficit and extent of neuronal necrosis attributable to middle cerebral artery occlusion in rats. Statistical validation. Stroke. 1995;26:627–34.CrossRefPubMed
42.
go back to reference Barber PA, Hoyte L, Colbourne F, Buchan AM. Temperature-regulated model of focal ischemia in the mouse: a study with histopathological and behavioral outcomes. Stroke. 2004;35:1720–5.CrossRefPubMed Barber PA, Hoyte L, Colbourne F, Buchan AM. Temperature-regulated model of focal ischemia in the mouse: a study with histopathological and behavioral outcomes. Stroke. 2004;35:1720–5.CrossRefPubMed
43.
go back to reference Schomacher M, Müller HD, Sommer C, Schwab S, Schäbitz WR. Endocannabinoids mediate neuroprotection after transient focal cerebral ischemia. Brain Res. 2008;1240:213–20.CrossRefPubMed Schomacher M, Müller HD, Sommer C, Schwab S, Schäbitz WR. Endocannabinoids mediate neuroprotection after transient focal cerebral ischemia. Brain Res. 2008;1240:213–20.CrossRefPubMed
44.
go back to reference Hara H, Friedlander RM, Gagliardini V, Ayata C, Fink K, Huang Z, et al. Inhibition of interleukin 1beta converting enzyme family proteases reduces ischemic and excitotoxic neuronal damage. Proc Natl Acad Sci U S A. 1997;94:2007–12.PubMedCentralCrossRefPubMed Hara H, Friedlander RM, Gagliardini V, Ayata C, Fink K, Huang Z, et al. Inhibition of interleukin 1beta converting enzyme family proteases reduces ischemic and excitotoxic neuronal damage. Proc Natl Acad Sci U S A. 1997;94:2007–12.PubMedCentralCrossRefPubMed
45.
go back to reference Schabitz WR, Li F, Irie K, Sandage Jr BW, Locke KW, Fisher M. Synergistic effects of a combination of low-dose basic fibroblast growth factor and citicoline after temporary experimental focal ischemia. Stroke. 1999;30:427–31. discussion 431-432.CrossRefPubMed Schabitz WR, Li F, Irie K, Sandage Jr BW, Locke KW, Fisher M. Synergistic effects of a combination of low-dose basic fibroblast growth factor and citicoline after temporary experimental focal ischemia. Stroke. 1999;30:427–31. discussion 431-432.CrossRefPubMed
46.
go back to reference Côté R, Battista RN, Wolfson C, Boucher J, Adam J, Hachinski V. The Canadian Neurological Scale: validation and reliability assessment. Neurology. 1989;39:638–43.CrossRefPubMed Côté R, Battista RN, Wolfson C, Boucher J, Adam J, Hachinski V. The Canadian Neurological Scale: validation and reliability assessment. Neurology. 1989;39:638–43.CrossRefPubMed
47.
go back to reference Folstein MF, Folstein SE, McHugh PR. “Mini-mental state”: a practical method for grading the cognitive state of patients for the clinician. J Psychiat Res. 1975;12:189–98.CrossRefPubMed Folstein MF, Folstein SE, McHugh PR. “Mini-mental state”: a practical method for grading the cognitive state of patients for the clinician. J Psychiat Res. 1975;12:189–98.CrossRefPubMed
48.
go back to reference Shah S, Cooper B. Documentation for measuring stroke rehabilitation outcomes. Aust Med Rec J. 1991;21:88–95. Shah S, Cooper B. Documentation for measuring stroke rehabilitation outcomes. Aust Med Rec J. 1991;21:88–95.
49.
go back to reference Trendelenburg G. Molecular regulation of cell fate in cerebral ischemia: role of the inflammasome and connected pathways. J Cereb Blood Flow Metab. 2014;34:1857–67.PubMedCentralCrossRefPubMed Trendelenburg G. Molecular regulation of cell fate in cerebral ischemia: role of the inflammasome and connected pathways. J Cereb Blood Flow Metab. 2014;34:1857–67.PubMedCentralCrossRefPubMed
50.
go back to reference Lai TW, Zhang S, Wang YT. Excitotoxicity and stroke: identifying novel targets for neuroprotection. Prog Neurobiol. 2014;115:157–88.CrossRefPubMed Lai TW, Zhang S, Wang YT. Excitotoxicity and stroke: identifying novel targets for neuroprotection. Prog Neurobiol. 2014;115:157–88.CrossRefPubMed
52.
go back to reference Burtrum D, Silverstein FS. Hypoxic-ischemic brain injury stimulates glial fibrillary acidic protein mRNA and protein expression in neonatal rats. Exp Neurol. 1994;126:112–8.CrossRefPubMed Burtrum D, Silverstein FS. Hypoxic-ischemic brain injury stimulates glial fibrillary acidic protein mRNA and protein expression in neonatal rats. Exp Neurol. 1994;126:112–8.CrossRefPubMed
53.
go back to reference Gehrmann J, Banati RB, Wiessner C, Hossmann KA, Kreutzberg GW. Reactive microglia in cerebral ischaemia: an early mediator of tissue damage? Neuropathol Appl Neurobiol. 1995;21:277–89.CrossRefPubMed Gehrmann J, Banati RB, Wiessner C, Hossmann KA, Kreutzberg GW. Reactive microglia in cerebral ischaemia: an early mediator of tissue damage? Neuropathol Appl Neurobiol. 1995;21:277–89.CrossRefPubMed
54.
go back to reference Walker EJ, Rosenberg GA. TIMP-3 and MMP-3 contribute to delayed inflammation and hippocampal neuronal death following global ischemia. Exp Neurol. 2009;216:122–31.PubMedCentralCrossRefPubMed Walker EJ, Rosenberg GA. TIMP-3 and MMP-3 contribute to delayed inflammation and hippocampal neuronal death following global ischemia. Exp Neurol. 2009;216:122–31.PubMedCentralCrossRefPubMed
55.
go back to reference Ahmad A, Genovese T, Impellizzeri D, Crupi R, Velardi E, Marino A, et al. Reduction of ischemic brain injury by administration of palmitoylethanolamide after transient middle cerebral artery occlusion in rats. Brain Res. 2012;1477:45–58.CrossRefPubMed Ahmad A, Genovese T, Impellizzeri D, Crupi R, Velardi E, Marino A, et al. Reduction of ischemic brain injury by administration of palmitoylethanolamide after transient middle cerebral artery occlusion in rats. Brain Res. 2012;1477:45–58.CrossRefPubMed
56.
go back to reference Han BH, Holtzman DM. BDNF protects the neonatal brain from hypoxic-ischemic injury in vivo via the ERK pathway. J Neurosci. 2000;20:5775–81.PubMed Han BH, Holtzman DM. BDNF protects the neonatal brain from hypoxic-ischemic injury in vivo via the ERK pathway. J Neurosci. 2000;20:5775–81.PubMed
57.
go back to reference Wang Y, Chang CF, Morales M, Chiang YH, Hoffer J. Protective effects of glial cell line-derived neurotrophic factor in ischemic brain injury. Ann NY Acad Sci. 2002;962:423–37.CrossRefPubMed Wang Y, Chang CF, Morales M, Chiang YH, Hoffer J. Protective effects of glial cell line-derived neurotrophic factor in ischemic brain injury. Ann NY Acad Sci. 2002;962:423–37.CrossRefPubMed
58.
go back to reference Mattila OS, Strbian D, Saksi J, Pikkarainen TO, Rantanen V, Tatlisumak T, et al. Cerebral mast cells mediate blood-brain barrier disruption in acute experimental ischemic stroke through perivascular gelatinase activation. Stroke. 2011;42:3600–5.CrossRefPubMed Mattila OS, Strbian D, Saksi J, Pikkarainen TO, Rantanen V, Tatlisumak T, et al. Cerebral mast cells mediate blood-brain barrier disruption in acute experimental ischemic stroke through perivascular gelatinase activation. Stroke. 2011;42:3600–5.CrossRefPubMed
59.
go back to reference Barnes PJ, Karin M. Nuclear factor-kappaB: a pivotal transcription factor in chronic inflammatory diseases. N Engl J Med. 1997;336:1066–71.CrossRefPubMed Barnes PJ, Karin M. Nuclear factor-kappaB: a pivotal transcription factor in chronic inflammatory diseases. N Engl J Med. 1997;336:1066–71.CrossRefPubMed
60.
go back to reference Bowie A, O’Neill LA. Oxidative stress and nuclear factor-kappaB activation: a reassessment of the evidence in the light of recent discoveries. Biochem Pharmacol. 2000;59:13–23.CrossRefPubMed Bowie A, O’Neill LA. Oxidative stress and nuclear factor-kappaB activation: a reassessment of the evidence in the light of recent discoveries. Biochem Pharmacol. 2000;59:13–23.CrossRefPubMed
62.
go back to reference Lerouet D, Beray-Berthat V, Palmier B, Plotkine M, Margaill I. Changes in oxidative stress, iNOS activity and neutrophil infiltration in severe transient focal cerebral ischemia in rats. Brain Res. 2002;958:166–75.CrossRefPubMed Lerouet D, Beray-Berthat V, Palmier B, Plotkine M, Margaill I. Changes in oxidative stress, iNOS activity and neutrophil infiltration in severe transient focal cerebral ischemia in rats. Brain Res. 2002;958:166–75.CrossRefPubMed
63.
go back to reference Tokime T, Nozaki K, Sugino T, Kikuchi H, Hashimoto N, Ueda K. Enhanced poly(ADP-ribosyl)ation after focal ischemia in rat brain. J Cereb Blood Flow Metab. 1998;18:991–7.CrossRefPubMed Tokime T, Nozaki K, Sugino T, Kikuchi H, Hashimoto N, Ueda K. Enhanced poly(ADP-ribosyl)ation after focal ischemia in rat brain. J Cereb Blood Flow Metab. 1998;18:991–7.CrossRefPubMed
64.
go back to reference Finucane DM, Bossy-Wetzel E, Waterhouse NJ, Cotter TG, Green DR. Bax-induced caspase activation and apoptosis via cytochrome c release from mitochondria is inhibitable by Bcl-xL. J Biol Chem. 1999;274:2225–33.CrossRefPubMed Finucane DM, Bossy-Wetzel E, Waterhouse NJ, Cotter TG, Green DR. Bax-induced caspase activation and apoptosis via cytochrome c release from mitochondria is inhibitable by Bcl-xL. J Biol Chem. 1999;274:2225–33.CrossRefPubMed
65.
go back to reference Kowaltowski AJ, Fenton RG, Fiskum G. Bcl-2 family proteins regulate mitochondrial reactive oxygen production and protect against oxidative stress. Free Radic Biol Med. 2004;37:1845–53.CrossRefPubMed Kowaltowski AJ, Fenton RG, Fiskum G. Bcl-2 family proteins regulate mitochondrial reactive oxygen production and protect against oxidative stress. Free Radic Biol Med. 2004;37:1845–53.CrossRefPubMed
66.
go back to reference Ahmad A, Crupi R, Impellizzeri D, Campolo M, Marino A, Esposito E, et al. Administration of palmitoylethanolamide (PEA) protects the neurovascular unit and reduces secondary injury after traumatic brain injury in mice. Brain Behav Immun. 2012;26:1310–21.CrossRefPubMed Ahmad A, Crupi R, Impellizzeri D, Campolo M, Marino A, Esposito E, et al. Administration of palmitoylethanolamide (PEA) protects the neurovascular unit and reduces secondary injury after traumatic brain injury in mice. Brain Behav Immun. 2012;26:1310–21.CrossRefPubMed
67.
go back to reference Citraro R, Russo E, Scicchitano F, van Rijn CM, Cosco D, Avagliano C, et al. Antiepileptic action of N-palmitoylethanolamine through CB1 and PPAR-α receptor activation in a genetic model of absence epilepsy. Neuropharmacology. 2013;69:115–1126.CrossRefPubMed Citraro R, Russo E, Scicchitano F, van Rijn CM, Cosco D, Avagliano C, et al. Antiepileptic action of N-palmitoylethanolamine through CB1 and PPAR-α receptor activation in a genetic model of absence epilepsy. Neuropharmacology. 2013;69:115–1126.CrossRefPubMed
68.
go back to reference Middleton Jr E, Kandaswami C, Theoharides TC. The effects of plant flavonoids on mammalian cells: implications for inflammation, heart disease, and cancer. Pharmacol Rev. 2000;52:673–751.PubMed Middleton Jr E, Kandaswami C, Theoharides TC. The effects of plant flavonoids on mammalian cells: implications for inflammation, heart disease, and cancer. Pharmacol Rev. 2000;52:673–751.PubMed
69.
go back to reference Seelinger G, Merfort I, Schempp CM. Anti-oxidant, anti-inflammatory and anti-allergic activities of luteolin. Planta Med. 2008;74:1667–77.CrossRefPubMed Seelinger G, Merfort I, Schempp CM. Anti-oxidant, anti-inflammatory and anti-allergic activities of luteolin. Planta Med. 2008;74:1667–77.CrossRefPubMed
70.
go back to reference Lovatel GA, Bertoldi K, Elsnerb VR, Piazza FV, Basso CG, Moysés Fdos S, et al. Long-term effects of pre and post-ischemic exercise following global cerebral ischemia on astrocyte and microglia functions in hippocampus from Wistar rats. Brain Res. 2014;1587:119–26.CrossRefPubMed Lovatel GA, Bertoldi K, Elsnerb VR, Piazza FV, Basso CG, Moysés Fdos S, et al. Long-term effects of pre and post-ischemic exercise following global cerebral ischemia on astrocyte and microglia functions in hippocampus from Wistar rats. Brain Res. 2014;1587:119–26.CrossRefPubMed
71.
go back to reference Yang Y, Salayandia VM, Thompson JF, Yang LY, Estrada EY, Yang Y. Attenuation of acute stroke injury in rat brain by minocycline promotes blood-brain barrier remodeling and alternative microglia/macrophage activation during recovery. J Neuroinflammation. 2015;12:26. doi:10.1186/s12974-015-0245-4.PubMedCentralCrossRefPubMed Yang Y, Salayandia VM, Thompson JF, Yang LY, Estrada EY, Yang Y. Attenuation of acute stroke injury in rat brain by minocycline promotes blood-brain barrier remodeling and alternative microglia/macrophage activation during recovery. J Neuroinflammation. 2015;12:26. doi:10.​1186/​s12974-015-0245-4.PubMedCentralCrossRefPubMed
72.
go back to reference Brifault C, Gras M, Liot D, May V, Vaudry D, Wurtz O. Delayed pituitary adenylate cyclase-activating polypeptide delivery after brain stroke improves functional recovery by inducing m2 microglia/macrophage polarization. Stroke. 2015;46(2):520–8. doi:10.1161/STROKEAHA.114.006864.CrossRefPubMed Brifault C, Gras M, Liot D, May V, Vaudry D, Wurtz O. Delayed pituitary adenylate cyclase-activating polypeptide delivery after brain stroke improves functional recovery by inducing m2 microglia/macrophage polarization. Stroke. 2015;46(2):520–8. doi:10.​1161/​STROKEAHA.​114.​006864.CrossRefPubMed
73.
go back to reference Strbian D, Kovanen PT, Karjalainen-Lindsberg ML, Tatlisumak T, Lindsberg PJ. An emerging role of mast cells in cerebral ischemia and hemorrhage. Ann Med. 2009;41(6):438–50.CrossRefPubMed Strbian D, Kovanen PT, Karjalainen-Lindsberg ML, Tatlisumak T, Lindsberg PJ. An emerging role of mast cells in cerebral ischemia and hemorrhage. Ann Med. 2009;41(6):438–50.CrossRefPubMed
74.
go back to reference Levy D, Edut S, Baraz-Goldstein R, Rubovitch V, Defrin R, Bree D, et al. Responses of dural mast cells in concussive and blast models of mild traumatic brain injury in mice: potential implications for post-traumatic headache. Cephalalgia. 2015. Levy D, Edut S, Baraz-Goldstein R, Rubovitch V, Defrin R, Bree D, et al. Responses of dural mast cells in concussive and blast models of mild traumatic brain injury in mice: potential implications for post-traumatic headache. Cephalalgia. 2015.
76.
go back to reference Anglemyer A, Horvath HT, Bero L. Healthcare outcomes assessed with observational study designs compared with those assessed in randomized trials. Cochrane Database Syst Rev. Issue 4. Art. No.: MR000034. doi:10.1002/14651858.MR000034.pub2. Anglemyer A, Horvath HT, Bero L. Healthcare outcomes assessed with observational study designs compared with those assessed in randomized trials. Cochrane Database Syst Rev. Issue 4. Art. No.: MR000034. doi:10.​1002/​14651858.​MR000034.​pub2.
77.
go back to reference Fornasari P, Montanari S. Utilizzo del protocollo di minima per l’ictus cerebrale (PMIC) per la valutazione dell’outcome e del percorso riabilitativo di un gruppo di pazienti ricoverati presso l’U.O. di medicina riabilitativa di Cesenatico. Eur Med Phys. 2008;44 (Suppl.1 to No.3). Fornasari P, Montanari S. Utilizzo del protocollo di minima per l’ictus cerebrale (PMIC) per la valutazione dell’outcome e del percorso riabilitativo di un gruppo di pazienti ricoverati presso l’U.O. di medicina riabilitativa di Cesenatico. Eur Med Phys. 2008;44 (Suppl.1 to No.3).
Metadata
Title
Co-ultramicronized Palmitoylethanolamide/Luteolin in the Treatment of Cerebral Ischemia: from Rodent to Man
Authors
Carlo Caltagirone
Carlo Cisari
Carlo Schievano
Rosanna Di Paola
Marika Cordaro
Giuseppe Bruschetta
Emanuela Esposito
Salvatore Cuzzocrea
Stroke Study Group
Publication date
01-02-2016
Publisher
Springer US
Published in
Translational Stroke Research / Issue 1/2016
Print ISSN: 1868-4483
Electronic ISSN: 1868-601X
DOI
https://doi.org/10.1007/s12975-015-0440-8

Other articles of this Issue 1/2016

Translational Stroke Research 1/2016 Go to the issue