Repositioning drugs for traumatic brain injury - N-acetyl cysteine and Phenserine

Barry J Hoffer, Chaim G Pick, Michael E Hoffer, Robert E Becker, Yung-Hsiao Chiang, Nigel H Greig, Barry J Hoffer, Chaim G Pick, Michael E Hoffer, Robert E Becker, Yung-Hsiao Chiang, Nigel H Greig

Abstract

Traumatic brain injury (TBI) is one of the most common causes of morbidity and mortality of both young adults of less than 45 years of age and the elderly, and contributes to about 30% of all injury deaths in the United States of America. Whereas there has been a significant improvement in our understanding of the mechanism that underpin the primary and secondary stages of damage associated with a TBI incident, to date however, this knowledge has not translated into the development of effective new pharmacological TBI treatment strategies. Prior experimental and clinical studies of drugs working via a single mechanism only may have failed to address the full range of pathologies that lead to the neuronal loss and cognitive impairment evident in TBI and other disorders. The present review focuses on two drugs with the potential to benefit multiple pathways considered important in TBI. Notably, both agents have already been developed into human studies for other conditions, and thus have the potential to be rapidly repositioned as TBI therapies. The first is N-acetyl cysteine (NAC) that is currently used in over the counter medications for its anti-inflammatory properties. The second is (-)-phenserine ((-)-Phen) that was originally developed as an experimental Alzheimer's disease (AD) drug. We briefly review background information about TBI and subsequently review literature suggesting that NAC and (-)-Phen may be useful therapeutic approaches for TBI, for which there are no currently approved drugs.

Keywords: N-acetyl cysteine; Phenserine; Traumatic brain injury.

Conflict of interest statement

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

Robert E. Becker does not have a competing interest but a conflict of interest. For disclosure, Robert E. Becker holds a patent on the use of phenserine in concussion/TBI and Alzheimer’s disease assigned to Aristea Translational Medicine Corporation of Utah. None of the other authors have any competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Figures

Fig. 1
Fig. 1
a Structure N-acetyl cysteine. b Post injury administration of NAC (50 mg/kg daily for 4 days) significantly improves MWM performance. MWM performance as measured by latency to reach the goal platform was compared between groups: TBI, TBI-NAC, and Sham. Both Sham and TBI-NAC groups have significantly shorter latencies to reach the goal platform as compared to the TBI group. Additionally, treatment with NAC after TBI improved performance in the MWM that reached sham levels. Data are presented as the mean ± SEM. *p < .05, ***p ≤ .001, sham relative to TBI. † p < .05 TBI-NAC relative to TBI. c Number of times animals crossed the within a 7.5 cm radius of the platform border during the probe trial. A one-way ANOVA showed significant differences between groups. Fisher’s LSD post hoc showed that sham and TBI-NAC (50 mg/kg daily for 4 days) had significantly better retention of the platform location as compared to TBI alone. Data are presented as the mean ± SEM. Brackets indicate comparisons between groups. *p < 0.05, **p < 0.01
Fig. 2
Fig. 2
Number of clinical symptoms at seven days
Fig. 3
Fig. 3
(−)-Phen is primarily metabolized by N-dethylation to yield (−)-N1- and (−)-N8-norphenserine that then are further N-dethylated to (−)-N1,N8-bisnorphenserine. Their brain/plasma ratios under steady-state conditions are shown
Fig. 4
Fig. 4
Human peripheral blood mononuclear cells (PBMCs) were isolated, from human blood, then cultured in the presence and absence of (−)-Phen for 24 h, and were then challenged with phytohemaggultanin (PHA: 3 μg/ml; Sigma-Aldrich) to induce inflammation and cytokine production. The detection limit for these assays is

Fig. 5

Cultured hippocampal neurons in triplicate…

Fig. 5

Cultured hippocampal neurons in triplicate were prepared from 18 to 20 day rat…

Fig. 5
Cultured hippocampal neurons in triplicate were prepared from 18 to 20 day rat (Sprague Dawley) embryos and were cultured for 7 days. They were treated with (−)-Phen (5 uM) followed by addition of an excitotoxic concentration of glutamate (50 uM). Neuronal viability was assessed 24 h after addition of glutamate (MTS assay). The results are plotted as percent neuronal survival ± SEM [58]

Fig. 6

Human SH-SY5Y were treated with…

Fig. 6

Human SH-SY5Y were treated with and without (−)-Phen and challenged with oxidative stress…

Fig. 6
Human SH-SY5Y were treated with and without (−)-Phen and challenged with oxidative stress (H2O2: 100 uM). Cell viability was quantified by MTS assay at 24 h. * designates comparisons with cells challenged with H2O2 (*p < 0.05, N ≥ 4 per group). Viability with (−)-Phen (30 uM) treatment was no different from control unchallenged cells

Fig. 7

a and b (−)-Phen (2.5…

Fig. 7

a and b (−)-Phen (2.5 mg/kg, i.p.) was administered for 21 consecutive days…
Fig. 7
a and b (−)-Phen (2.5 mg/kg, i.p.) was administered for 21 consecutive days to rats and mice. Animals were killed within 2 h of their final (−)-Phen dose or saline; a brain (cortex) sample was taken and immediately frozen to –70oC and thereafter analysed for Aβ by ELISA. (−)-Phen significantly (p < 0.05) lowered APP, Aβ (1-40 and 1-42) levels vs. controls. This decline, particularly in Aβ42 levels, was likewise found in wild type mice dosed with (−)-Phen (2.5 mg/kg and 7.5 mg/kg, i.p.) for 21 days [80]

Fig. 8

Administration of (−)-Phen to humans,…

Fig. 8

Administration of (−)-Phen to humans, by gradually escalating the dose to achieve 15…

Fig. 8
Administration of (−)-Phen to humans, by gradually escalating the dose to achieve 15 mg BID resulted in a decline (~20%) in levels of Aβ42 evaluated in the plasma time-dependently following the final (−)-Phen dose. This reduction coincided with the achieval of peak plasma (−)-Phen concentrations and suggests that the maintenance of long-term steady-state levels of (−)-Phen by slow-release formulations could provide a sustained lowering of Aβ42 in humans [83]. Phenserine tartrate was administered orally to healthy volunteers. Days 1-28: 10 mg BID; Days 29-34: 15 BID; Day 35: 15 mg. Blood samples were drawn on Days 1, 28, 29 and 35. Plasma samples were analyzed for Aβ1-42 using a sandwich ELISA. Plasma phenserine concentrations were determined by LC/MS/MS

Fig. 9

Primary SVZ progenitors cells were…

Fig. 9

Primary SVZ progenitors cells were isolated from the lateral and medial ganglionic eminence…

Fig. 9
Primary SVZ progenitors cells were isolated from the lateral and medial ganglionic eminence of mouse embryos at embryonic day E13.5, and following trituration to a single cell suspension were grown as neurospheres for day 6 or 7 in vitro in the presence and absence of (−)-Phen analogs (0.01 μM) – which increased cell survival [78, 104]

Fig. 10

a and b . A…

Fig. 10

a and b . A time-dependent plasma AChE inhibition achieved by (−)-Phen in…
Fig. 10
a and b. A time-dependent plasma AChE inhibition achieved by (−)-Phen in anesthetized rats following a single dose, in which cholinesterase inhibition was achieved by the combined action of (−)-Phen and its primary metabolites. (−)-Phen and active metabolites readily enter the brain (see Fig. 3), and thereby induce brain AChE inhibition and elevate acetylcholine levels [110]. By contrast, (−)-Physostigmine at a higher dose achieves lower plasma AChE inhibition, has less brain uptake than (−)-Phen, is short-lived in vivo, and is associated with greater adverses actions [72]. b: Time-dependent plasma AChE inhibition and predicted brain pharmacokinetics of (−)-Phen and primary metabolites in humans after single acute dosing

Fig. 11

mild TBI mice demonstrate a…

Fig. 11

mild TBI mice demonstrate a deficit in visual memory compared with control uninjured…

Fig. 11
mild TBI mice demonstrate a deficit in visual memory compared with control uninjured (Sham) animals (**p < 0.01), in which (−)-Phen administration significantly ameliorated (at both doses **p < 0.01 vs. mTBI alone) [58]. A washout period of 2 days before cognitive evaluation ensured no confound in relation to any direct action to improve cognition. These data are thus interpreted as evidence for an effect of Phen against post injury pathology allowing reduced cognitive deficits in (−)-Phen treated animals (mTBI: mild TBI, Phen: (−)-Phen)

Fig. 12

Y-Maze was assessed two days…

Fig. 12

Y-Maze was assessed two days after (−)-Phen washout, evaluating two clinically translatable doses…

Fig. 12
Y-Maze was assessed two days after (−)-Phen washout, evaluating two clinically translatable doses (2.5 and 5.0 mg/kg BID × 5 days) initiated post mTBI. Whereas mTBI challenged mice demonstrate a significant deficit in spatial memory vs. control uninjured (Sham) animals (**p < 0.01). (−)-Phen administration significantly ameliorated this deficit (##p < 0.01 for 2.5 mg/kg and #p < 0.05 for 5 mg/kg vs. mTBI alone) [58]. Likewise, these data are interpreted as evidence for a positive effect against post injury pathology allowing reduced cognitive deficits in (−)-Phen treated animals (mTBI: mild TBI, Phen: (−)-Phen)
All figures (12)
Fig. 5
Fig. 5
Cultured hippocampal neurons in triplicate were prepared from 18 to 20 day rat (Sprague Dawley) embryos and were cultured for 7 days. They were treated with (−)-Phen (5 uM) followed by addition of an excitotoxic concentration of glutamate (50 uM). Neuronal viability was assessed 24 h after addition of glutamate (MTS assay). The results are plotted as percent neuronal survival ± SEM [58]
Fig. 6
Fig. 6
Human SH-SY5Y were treated with and without (−)-Phen and challenged with oxidative stress (H2O2: 100 uM). Cell viability was quantified by MTS assay at 24 h. * designates comparisons with cells challenged with H2O2 (*p < 0.05, N ≥ 4 per group). Viability with (−)-Phen (30 uM) treatment was no different from control unchallenged cells
Fig. 7
Fig. 7
a and b (−)-Phen (2.5 mg/kg, i.p.) was administered for 21 consecutive days to rats and mice. Animals were killed within 2 h of their final (−)-Phen dose or saline; a brain (cortex) sample was taken and immediately frozen to –70oC and thereafter analysed for Aβ by ELISA. (−)-Phen significantly (p < 0.05) lowered APP, Aβ (1-40 and 1-42) levels vs. controls. This decline, particularly in Aβ42 levels, was likewise found in wild type mice dosed with (−)-Phen (2.5 mg/kg and 7.5 mg/kg, i.p.) for 21 days [80]
Fig. 8
Fig. 8
Administration of (−)-Phen to humans, by gradually escalating the dose to achieve 15 mg BID resulted in a decline (~20%) in levels of Aβ42 evaluated in the plasma time-dependently following the final (−)-Phen dose. This reduction coincided with the achieval of peak plasma (−)-Phen concentrations and suggests that the maintenance of long-term steady-state levels of (−)-Phen by slow-release formulations could provide a sustained lowering of Aβ42 in humans [83]. Phenserine tartrate was administered orally to healthy volunteers. Days 1-28: 10 mg BID; Days 29-34: 15 BID; Day 35: 15 mg. Blood samples were drawn on Days 1, 28, 29 and 35. Plasma samples were analyzed for Aβ1-42 using a sandwich ELISA. Plasma phenserine concentrations were determined by LC/MS/MS
Fig. 9
Fig. 9
Primary SVZ progenitors cells were isolated from the lateral and medial ganglionic eminence of mouse embryos at embryonic day E13.5, and following trituration to a single cell suspension were grown as neurospheres for day 6 or 7 in vitro in the presence and absence of (−)-Phen analogs (0.01 μM) – which increased cell survival [78, 104]
Fig. 10
Fig. 10
a and b. A time-dependent plasma AChE inhibition achieved by (−)-Phen in anesthetized rats following a single dose, in which cholinesterase inhibition was achieved by the combined action of (−)-Phen and its primary metabolites. (−)-Phen and active metabolites readily enter the brain (see Fig. 3), and thereby induce brain AChE inhibition and elevate acetylcholine levels [110]. By contrast, (−)-Physostigmine at a higher dose achieves lower plasma AChE inhibition, has less brain uptake than (−)-Phen, is short-lived in vivo, and is associated with greater adverses actions [72]. b: Time-dependent plasma AChE inhibition and predicted brain pharmacokinetics of (−)-Phen and primary metabolites in humans after single acute dosing
Fig. 11
Fig. 11
mild TBI mice demonstrate a deficit in visual memory compared with control uninjured (Sham) animals (**p < 0.01), in which (−)-Phen administration significantly ameliorated (at both doses **p < 0.01 vs. mTBI alone) [58]. A washout period of 2 days before cognitive evaluation ensured no confound in relation to any direct action to improve cognition. These data are thus interpreted as evidence for an effect of Phen against post injury pathology allowing reduced cognitive deficits in (−)-Phen treated animals (mTBI: mild TBI, Phen: (−)-Phen)
Fig. 12
Fig. 12
Y-Maze was assessed two days after (−)-Phen washout, evaluating two clinically translatable doses (2.5 and 5.0 mg/kg BID × 5 days) initiated post mTBI. Whereas mTBI challenged mice demonstrate a significant deficit in spatial memory vs. control uninjured (Sham) animals (**p < 0.01). (−)-Phen administration significantly ameliorated this deficit (##p < 0.01 for 2.5 mg/kg and #p < 0.05 for 5 mg/kg vs. mTBI alone) [58]. Likewise, these data are interpreted as evidence for a positive effect against post injury pathology allowing reduced cognitive deficits in (−)-Phen treated animals (mTBI: mild TBI, Phen: (−)-Phen)

References

    1. Hyder AA, Wunderlich CA, Puvanachandra P, Gururaj G, Ko-busingye OC. The impact of traumatic brain injuries: a global perspective. Neuro Rehabilitation. 2007;22:341–353.
    1. Ruff RL, Riechers RG. Effective treatment of traumatic brain injury: learning from experience. JAMA. 2012;308:2032–2033. doi: 10.1001/jama.2012.14008.
    1. Langlois JA, Rutland-Brown W, Wald MM. The epidemiology and impact of traumatic brain injury: a brief overview. J Head Trauma Rehabil. 2006;21:375–378. doi: 10.1097/00001199-200609000-00001.
    1. Prins ML, Giza CC. Repeat traumatic brain injury in the developing brain. Int J Dev Neurosci. 2012;30:185–190. doi: 10.1016/j.ijdevneu.2011.05.009.
    1. Tagliaferri F, Compagnone C, Korsic M, Servadei F, Kraus J. Systematic review of brain injury epidemiology in Europe. Acta Neurochir. 2006;148:255–268. doi: 10.1007/s00701-005-0651-y.
    1. Shi HY, Hwang SL, Lee KT, Lin CL. Temporal trends and volume-outcome associations after traumatic brain injury: a 12- year study in Taiwan. Clinical article J Neurosurg. 2013;118:732–738. doi: 10.3171/2012.12.JNS12693.
    1. Gardner RC, Burke JF, Nettiksimmons J, Kaup A, Barnes DE, Yaffe K. Dementia risk after traumatic brain injury vs nonbrain trauma: the role of age and severity. JAMA Neurol. 2014;71(12):1490–1497. doi: 10.1001/jamaneurol.2014.2668.
    1. Barnes DE, Kaup A, Kirby KA, Byers AL, Diaz-Arrastia R, Yaffe K. Traumatic brain injury and risk of dementia in older veterans. Neurology. 2014;83(4):312–319. doi: 10.1212/WNL.0000000000000616.
    1. Greig NH, Sambamurti K, Lahiri DK, Becker RE. Amyloid-β precursor protein synthesis inhibitors for Alzheimer's disease treatment. Ann Neurol. 2014;76:629–630. doi: 10.1002/ana.24254.
    1. Tweedie D, Rachmany L, Rubovitch V, Zhang Y, Becker KG, Perez E, et al. Changes in mouse cognition and hippocampal gene expression observed in a mild physical- and blast-traumatic brain injury. Neurobiol Dis. 2013;54:1–11. doi: 10.1016/j.nbd.2013.02.006.
    1. Tweedie D, Rachmany L, Rubovitch V, Lehrmann E, Zhang Y, Becker KG, et al. Exendin-4, a glucagon-like peptide-1 receptor agonist prevents mTBI-induced changes in hippocampus gene expression and memory deficits in mice. Exp Neurol. 2013;239:170–182. doi: 10.1016/j.expneurol.2012.10.001.
    1. Goldstein LE, Fisher AM, Tagge CA, et al. Chronic traumatic encephalopathy in blast-exposed military veterans and a blast neurotrauma mouse model. Sci Transl Med. 2012;4(134):134ra60. doi: 10.1126/scitranslmed.3003716.
    1. LaPlaca MC, Simon CM, Prado GR, Cullen DK. CNS injury biomechanics and experimental models. Prog Brain Res. 2007;161:13–26. doi: 10.1016/S0079-6123(06)61002-9.
    1. Barkhoudarian G, Hovda DA, Giza C. The molecular pathophysiology of concussive brain injury. Clin Sports Med. 2011;30:33–48. doi: 10.1016/j.csm.2010.09.001.
    1. Greve MW, Zink BJ. Pathophysiology of traumatic brain injury. Mt Sinai J Med. 2009;76(2):97–104.
    1. Bachstetter AD, Norris CM, Sompol P, Wilcock DM, Goulding D, Neltner JH, et al. Early stage drug treatment that normalizes proinflammatory cytokine production attenuates synaptic dysfunction in a mouse model that exhibits age-dependent progression of Alzheimer's disease-related pathology. J Neurosci. 2012;32:10201–10. doi:10.1523/JNEUROSCI.1496-12.2012.
    1. Schmidt OI, Heyde CE, Ertel W, Stahel PF. Closed head injury - an inflammatory disease? Brain Res Rev. 2005;48:388–399. doi: 10.1016/j.brainresrev.2004.12.028.
    1. Moppett IK. Traumatic brain injury: assessment, resuscitation and early management. Br J Anaesth. 2007;99:18–31. doi: 10.1093/bja/aem128.
    1. Chen G, Shi J, Hu Z, Hang C. Inhibitory effect on cerebral inflammatory response following traumatic brain injury in rats: a potential neuroprotective mechanism of N-acetylcysteine. Mediat Inflamm. 2008;2008:716458. doi:10.1155/2008/716458.
    1. Ellis EF, Dodson LY, Police RJ. Restoration of cerebrovascular responsiveness to hyperventilation by the oxygen radical scavenger n-acetylcysteine. J Neurosurg. 1991;75(5):774–779. doi: 10.3171/jns.1991.75.5.0774.
    1. Bergold P, Haber M, Dash P, Grill R, Grin’kina N, Abdel-Baki S. Minocycline and N-Acetlycysteine modulates neuroinflammation and produces remyelination following controlled cortical impact. J Neurotrauma. 2012;29:A109–A110.
    1. Hicdonmez T, Kanter M, Tiryaki M, Parsak T, Cobanoglu S. Neuroprotective effects of N-acetylcysteine on experimental closed head trauma in rats. Neurochem Res. 2006;31:473–448. doi: 10.1007/s11064-006-9040-z.
    1. Khan M, Sekhon B, Jatana M, Giri S, Gilg AG, Sekhon C, Singh I, Singh AK. Administration of Nacetylcysteine after focal cerebral ischemia protects brain and reduces inflammation in a rat model of experimental stroke. J Res. 2004;76:519–527.
    1. Sekhon B, Sekhon C, Khan M, Patel SJ, Singh I, Singh AK. N-acetyl cysteine protects against injury in a rat model of focal cerebral ischemia. Brain Res. 2003;971:1–8. doi: 10.1016/S0006-8993(03)02244-3.
    1. Gilgun-Sherki Y, Rosenbaum Z, Melamed E, Offen D. Antioxidant therapy in acute central nervous system injury: current state. Pharmacol Rev. 2002;54:271–284. doi: 10.1124/pr.54.2.271.
    1. Santangelo F. Intracellular thiol concentration modulating inflammatory response: influence on the regulation of cell functions through cysteine prodrug approach. Curr Med Chem. 2003;10:2599–2610. doi: 10.2174/0929867033456567.
    1. Pahan K, Sheikh FG, Namboodiri AMS, Singh I. Nacetylcysteine inhibits induction of NO production by endotoxin or cytokine stimulated rat peritoneal macrophages, C6 glial cells and astrocytes. Free Radic Biol Med. 1998;24:39–48. doi: 10.1016/S0891-5849(97)00137-8.
    1. Lappas M, Permezel M, Rice GE. N-acetyl-cysteine inhibits phospholipid metabolism, proinflammatory cytokine release, protease activity, and nuclear factor-kappaB deoxyribonucleic acid-binding activity in human fetal membranes in vitro. J Clin Endocrinol Metab. 2003;88(4):1723–1729. doi: 10.1210/jc.2002-021677.
    1. Cuzzocrea S, Mazzon E, Costantino G, Serraino I, Dugo L, Calabro G, et al. Beneficial effects of nacetylcysteine on ischaemic brain injury. Br J Pharmacol. 2000;130:1219–1226. doi: 10.1038/sj.bjp.0703421.
    1. Tsai SY, Hayashi T, Harvey BK, Wang Y, Wu WW, Shen RF, et al. Sigma-1 receptors regulate hippocampal dendritic spine formation via a free radical-sensitive mechanism involving Rac1•GTP pathway. PNAS. 2009;106(52):22468-73.
    1. Eakin K, Baratz-Goldstein R, Pick CG, Zindel O, Balaban CD, Hoffer ME, et al. Efficacy of N-acetyl cysteine in traumatic brain injury. PLoS One. 2014;9(4):e90617. doi: 10.1371/journal.pone.0090617.
    1. Farr SA, Poon HF, Dogrukol-Ak D, Drake J, Banks WA, Eyerman E, et al. The antioxidants alpha-lipoic acid and N-acetylcysteine reverse memory impairment and brain oxidative stress in aged SAMP8 mice. J Neurochem. 2003;84:1173–1183. doi: 10.1046/j.1471-4159.2003.01580.x.
    1. Lanté F, Meunier J, Guiramand J, De Jesus Ferreira MC, Cambonie G, Aimar R, et al. Late N-acetylcysteine treatment prevents the deficits induced in the offspring of dams exposed to an immune stress during gestation. Hippocampus. 2008;18:602–609. doi: 10.1002/hipo.20421.
    1. Miguel J, Kulak A, Gholam-Razaee MM, Cuenod M, Gruetter R, Do KQ. N-Acetylcysteine normalizes Neurochemical changes in the glutathione-deficient schizophrenia mouse model during development. Biol Psychiatry. 2012;71:1006–1014. doi: 10.1016/j.biopsych.2011.07.035.
    1. Roth TL, Nayak D, Atanasijevic T, Koretsky AP, Latour LL, McGavern DB. Transcranial amelioration of inflammation and cell death after brain injury. Nature. 2014;505:223–228. doi: 10.1038/nature12808.
    1. Moussawi K, Pacchioni A, Moran M, Olive MF, Gass JT, Lavin A, et al. NAcetylcysteine reverses cocaine-induced metaplasticity. Nat Neurosci. 2009;12:182–189. doi: 10.1038/nn.2250.
    1. Hoffer ME, Balaban C, Slade MD, Tsao JW, Hoffer B. Amelioration of acute sequelae of blast induced mild traumatic brain injury by N-acetyl cysteine: a double-blind, placebo controlled study. PLoS One. 2013;8:e54163. doi: 10.1371/journal.pone.0054163.
    1. Diaz-Arrastia R, Kochanek PM, Bergold P, Kenney K, Marx C, Grimes J, et al. Pharmacotherapy of traumatic brain injury: state of the science and the road forward report of the Department of Defense Neurotrauma Pharmacology Workgroup. J Neurotrauma. 2014;31(2):135–58. doi:10.1089/neu.2013.3019.
    1. Verbois SL, Scheff SW, Pauly JR. Time-dependent changes in rat brain cholinergic receptor expression after experimental brain injury. J Neurotrauma. 2002;19:1569–1585. doi: 10.1089/089771502762300238.
    1. Murdoch I, Perry EK, Court JA, Graham DI, Dewar D. Cortical cholinergic dysfunction after human head injury. J Neurotrauma. 1998;15:295–305. doi: 10.1089/neu.1998.15.295.
    1. Dewar D, Graham DI. Depletion of choline acetyltransferase activity but preservation of M1 and M2 muscarinic receptor binding sites in temporal cortex following head injury: a preliminary human post-mortem study. J Neurotrauma. 1996;13:181–187.
    1. Schmidt RH, Grady MS. Loss of forebrain cholinergic neurons following fluid-percussion injury: implications for cognitive impairment in closed head injury. J Neurosurg. 1995;83:496–502. doi: 10.3171/jns.1995.83.3.0496.
    1. Poole NA, Agrawal N. Cholinomimetic agents and neurocognitive impairment following head injury: a systematic review. Brain Inj. 2008;22:519–534. doi: 10.1080/02699050802132495.
    1. Holschneider DP, Guo Y, Roch M, Norman KM, Scremin OU. Acetylcholinesterase inhibition and locomotor function after motor-sensory cortex impact injury. J Neurotrauma. 2011;28:1909–1919. doi: 10.1089/neu.2011.1978.
    1. Shaw KE, Bondi CO, Light SH, Massimino LA, McAloon RL, Monaco CM, Kline AE. Donepezil is ineffective in promoting motor and cognitive benefits after controlled cortical impact injury in male rats. J Neurotrauma. 2013;30:557–564. doi: 10.1089/neu.2012.2782.
    1. Maas AIR, Stocchetti N, Bullock R. Moderate and severe traumatic brain injury in adults. Lancet Neurol. 2008;7:728–741. doi: 10.1016/S1474-4422(08)70164-9.
    1. Cornelius C, Crupi R, Calabrese V, Graziano A, Milone P, Pennisi G, et al. Traumatic brain injury: oxidative stress and neuroprotection. Antioxid Redox Signal. 2013;19:836–853. doi: 10.1089/ars.2012.4981.
    1. Bornstein MB. Presence and action of acetylcholine in experimental brain trauma. J Neurophysiol. 1946;9:349–366.
    1. Sachs E., Jr Acetylcholine and serotonin in the spinal fluid. J Neurosurg. 1957;14:22–27. doi: 10.3171/jns.1957.14.1.0022.
    1. Metz B. Acetylcholine and experimental brain injury. J Neurosurg. 1971;35:523–528. doi: 10.3171/jns.1971.35.5.0523.
    1. Lyeth BG, Jiang JY, Robinson SE, Guo H, Jenkins LW. Hypothermia blunts acetylcholine increase in CSF of traumatically brain injured rats. Mol Chem Neuropathol. 1993;18:247–256. doi: 10.1007/BF03160117.
    1. Tower DB, McEachern D. Acetylcholine and neuronal activity. I. Cholinesterase patterns and acetylcholine in the cerebrospinal fluid of patients with craniocerebral trauma. Canad J Biochem. 1949;27:105–119.
    1. Ward AA. Jr Atropine in the treatment of closed head injury. J Neurosurg;1950;7: 398–402.
    1. Ruge D. The use of cholinergic blocking agents in the treatment of cranio-cerebral injuries. J Neurosurg. 1954;11:77–83. doi: 10.3171/jns.1954.11.1.0077.
    1. Lechner H. Anticholinergic therapy of craniocerebral trauma. Wien Klin Wochenschr. 1963;75:560.
    1. Lyeth BG, Liu S, Hamm RJ. Combined scopolamine and morphine treatment of traumatic brain injury in the rat. Brain Res. 1993;617:69–75. doi: 10.1016/0006-8993(93)90614-S.
    1. Cox CD, West EJ, Liu MC, Wang KK, Hayes RL, Lyeth BG. Dicyclomine, an M1 muscarinic antagonist, reduces biomarker levels, but not neuronal degeneration, in fluid percussion brain injury. J Neurotrauma. 2008;25:1355–1365. doi: 10.1089/neu.2008.0671.
    1. Tweedie D, Fukui K, Li Y, Yu QS, Barak S, Tamargo IA, et al. Cognitive impairments induced by concussive mild traumatic brain injury in mouse are ameliorated by treatment with Phenserine via multiple non-cholinergic and cholinergic mechanisms. PLoS One. 2016;11(6):e0156493. doi: 10.1371/journal.pone.0156493.
    1. Rachmany L, Tweedie D, Rubovitch V, Yu QS, Li Y, Wang JY, et al. Cognitive impairments accompanying rodent mild traumatic brain injury involve p53-dependent neuronal cell death and are ameliorated by the tetrahydrobenzothiazole PFT-α. PLoS One. 2013;8:e79837. doi: 10.1371/journal.pone.0079837.
    1. Zohar O, Schreiber S, Getslev V, Schwartz JP, Mullins PG, Pick CG. Closed-head minimal traumatic brain injury produces long-term cognitive deficits in mice. Neuroscience. 2003;118:949–955. doi: 10.1016/S0306-4522(03)00048-4.
    1. Zohar O, Rubovitch V, Milman A, Schreiber S, Pick CG. Behavioral consequences of minimal traumatic brain injury in mice. Acta Neurobiol Exp. 2011;71:36–45.
    1. Milman A, Rosenberg A, Weizman R, Pick CG. Mild traumatic brain injury induces persistent cognitive deficits and behavioral disturbances in mice. J Neurotrauma. 2005;22:1003–1010. doi: 10.1089/neu.2005.22.1003.
    1. Baratz R, Tweedie D, Rubovitch V, Luo W, Yoon JS, Hoffer BJ, et al. Tumor necrosis factor-α synthesis inhibitor, 3,6′-dithiothalidomide, reverses behavioral impairments induced by minimal traumatic brain injury in mice. J Neurochem. 2010;118:1032–1042. doi: 10.1111/j.1471-4159.2011.07377.x.
    1. Greig NH, Tweedie D, Rachmany L, Li Y, Rubovitch V, Schreiber S, Chiang YH, et al. Incretin mimetics as pharmacologic tools to elucidate and as a new drug strategy to treat traumatic brain injury. Alzheimers Dement. 2014;10(1 Suppl):S62–S75. doi: 10.1016/j.jalz.2013.12.011.
    1. Gardner RC, Yaffe K. Epidemiology of mild traumatic brain injury and neurodegenerative disease. Mol Cell Neurosci. 2015;66(Pt B):75–80. doi: 10.1016/j.mcn.2015.03.001.
    1. Crane PK, Gibbons LE, Dams-O'Connor K, Trittschuh E, Leverenz JB, Keene CD, et al. Association of Traumatic Brain Injury with Late-Life Neurodegenerative Conditions and Neuropathologic Findings. JAMA Neurol. 2016;73:1062–1069. doi: 10.1001/jamaneurol.2016.1948.
    1. Young JS, Hobbs JG, Bailes JE. The impact of traumatic brain injury on the aging brain. Curr Psychiatry Rep. 2016;18:81. doi: 10.1007/s11920-016-0719-9.
    1. Reale M, Di Nicola M, Velluto L, D'Angelo C, Costantini E, Lahiri DK, et al. Selective acetyl- and butyrylcholinesterase inhibitors reduce amyloid-β ex vivo activation of peripheral chemo-cytokines from Alzheimer's disease subjects: exploring the cholinergic anti-inflammatory pathway. Curr Alzheimer Res. 2014;11:608–622. doi: 10.2174/1567205010666131212113218.
    1. Lozano D, Gonzales-Portillo GS, Acosta S, de la Pena I, Tajiri N, Kaneko Y, et al. Neuroinflammatory responses to traumatic brain injury: etiology, clinical consequences, and therapeutic opportunities. Neuropsychiatr. Dis. Treat. 2015;11:97–106.
    1. Finnie JW. Neuroinflammation: beneficial and detrimental effects after traumatic brain injury. Inflammopharmacology. 2013;21:309–320. doi: 10.1007/s10787-012-0164-2.
    1. Greig NH, Pei XF, Soncrant TT, Ingram DK, Brossi A. Phenserine and ring C hetero-analogues: drug candidates for the treatment of Alzheimer's disease. Med Res Rev. 1995;15:3–31. doi: 10.1002/med.2610150103.
    1. Greig NH, Ruckle J, Comer P, Brownell L, Holloway HW, Flanagan DR Jr, et al. Anticholinesterase and pharmacokinetic profile of phenserine in healthy elderly human subjects. Curr Alzheimer Res 2005;2:483-492.
    1. Winblad B, Giacobini E, Frölich L, Friedhoff LT, Bruinsma G, Becker RE, et al. Phenserine efficacy in Alzheimer's disease. J Alzheimers Dis. 2010;22:1201–1208. doi: 10.3233/JAD-2010-101311.
    1. Yu Q, Greig NH, Holloway HW, Brossi A. Syntheses and anticholinesterase activities of (3aS)-N1, N8-bisnorphenserine, (3aS)-N1,N8-bisnorphysostigmine, their antipodal isomers, and other potential metabolites of phenserine. J Med Chem. 1998;41:2371–2379. doi: 10.1021/jm9800494.
    1. Yu QS, Reale M, Kamal MA, Holloway HW, Luo W, Sambamurti K, et al. Synthesis of the Alzheimer drug Posiphen into its primary metabolic products (+)-N1-norPosiphen, (+)-N8-norPosiphen and (+)-N1, N8-bisnorPosiphen, their inhibition of amyloid precursor protein, α-Synuclein synthesis, interleukin-1β release, and cholinergic action. Antinflamm Antiallergy Agents Med Chem. 2013;12:117–128. doi: 10.2174/1871523011312020003.
    1. Frankola KA, Greig NH, Luo W, Tweedie D. Targeting TNF-α to elucidate and ameliorate neuroinflammation in neurodegenerative diseases. CNS Neurol Disord Drug Targets. 2011;10:391–403. doi: 10.2174/187152711794653751.
    1. Chen YC, Mao H, Yang KH, Abel T, Meaney DF. A controlled cortical impact technique of model mild traumatic brain injury mechanics in mice. Front Neruol. 2014;5:100.
    1. Lilja AM, Luo Y, Yu QS, Röjdner J, Li Y, Marini AM, et al. Neurotrophic and neuroprotective actions of (−)- and (+)-phenserine, candidate drugs for Alzheimer’s disease. PLoS One. 2013;8(1):e54887. doi: 10.1371/journal.pone.0054887.
    1. Shaw KT, Utsuki T, Rogers J, Yu QS, Sambamurti K, Brossi A. Phenserine regulates translation of beta -amyloid precursor protein mRNA by a putative interleukin-1 responsive element, a target for drug development. PNAS. 2001;98:7605–7610. doi: 10.1073/pnas.131152998.
    1. Lahiri DK, Chen D, Maloney B, Holloway HW, Yu QS, Utsuki T, et al. The experimental Alzheimer’s disease drug Posiphen lowers amyloid–beta peptide levels in cell culture and mice. J Pharmacol Exp Ther. 2007;320:386–396. doi: 10.1124/jpet.106.112102.
    1. Mikkilineni S, Cantuti-Castelvetri I, Cahill CM, Balliedier A, Greig NH, Rogers JT. The anticholinesterase phenserine and its enantiomer posiphen as 5'untranslated-region-directed translation blockers of the Parkinson's alpha synuclein expression. Parkinsons Dis. 2012;142372
    1. Haroutunian V, Greig N, Pei XF, Utsuki T, Gluck R, Acevedo LD, et al. Pharmacological modulation of Alzheimer's beta-amyloid precursor protein levels in the CSF of rats with forebrain cholinergic system lesions. Brain Res Mol Brain Res. 1997;46:161–168. doi: 10.1016/S0169-328X(96)00297-5.
    1. Sambamurti K, Prakasam A, Anitha S, Venugopa C, Cullen E, Zhou Y, et al. Oral phenserine tartrate treatment is associated with reduced plasma Ab 1-42 in a phase I clinical study. In, Alzheimer's disease new advances (Khalid Iqbal K, Winblad B, Avila J). Bologna: Medimond, via Maserati 5; 2007. p. 571–5.
    1. Friedlich AL, Tanzi RE, Rogers JT. The 5′-untranslated region of Parkinson's disease alpha-synuclein messenger RNA contains a predicted iron responsive element. Mol Psychiatry. 2007;12:222–223. doi: 10.1038/sj.mp.4001937.
    1. Olivares D, Huang X, Branden L, Greig NH, Rogers JT. Physiological and pathological role of alpha-synuclein in Parkinson's disease through iron mediated oxidative stress; the role of a putative iron-responsive element. Int J Mol Sci. 2009;10:1226–1260. doi: 10.3390/ijms10031226.
    1. Cahill CM, Lahiri DK, Huang X, Rogers JT. Amyloid precursor protein and alpha synuclein translation, implications for iron and inflammation in neurodegenerative diseases. Biochim Biophys Acta. 2009;1790:615–628. doi: 10.1016/j.bbagen.2008.12.001.
    1. Smith DH, Chen X, Iwata A, Graham DI. Amyloid beta accumulation in axons after traumatic brain injury in humans. J Neurosurg. 2003;98:1072–1077. doi: 10.3171/jns.2003.98.5.1072.
    1. Van Den Heuvel C, Finnie JW, Blumbergs PC, Manavis J, Jones NR, Reilly PL, et al. Upregulation of neuronal amyloid pre- cursor protein (APP) and APP mRNA following magnesium sulphate (MgSO4) therapy in trau- matic brain injury. Neurotrauma. 2000;17:1041–1053. doi: 10.1089/neu.2000.17.1041.
    1. Luth H, Holzer M, Gartner U, Staufenbiel M, Arendt T. Expression of endothelial and inducible NOS-isoforms is increased in Alzheimer’s disease, in APP23 transgenic mice and after experimental brain lesion in rat: evidence for an induction by amyloid pathology. Brain Res. 2001;913:57–67. doi: 10.1016/S0006-8993(01)02758-5.
    1. Iwata A, Chen X, McIntosh TK, Browne K, Smith D. Long-term accumulation of amyloid-beta in axons following brain trauma without persistent upregulation of amyloid pre- cursor protein genes. J Neuropathol Exp Neurol. 2002;61:1056–1068. doi: 10.1093/jnen/61.12.1056.
    1. Smith DH, Chen XH, Nonaka M, Trojanowski JQ, Lee VM, Saatman KE, Leoni MJ, Xu BN, Wolf JA, Meaney DF. Accumulation of amyloid and tau and the formation of neurofilament inclusions following diffuse brain injury in the pig. J Neuropathol Exp Neurol. 1999;58:982–992. doi: 10.1097/00005072-199909000-00008.
    1. Lahiri DK, Maloney B, Zawia NH. The LEARn model: an epigenetic explanation for idiopathic neurobiological diseases. Mol Psychiatry. 2009;14:992–1003. doi: 10.1038/mp.2009.82.
    1. Maloney B, Lahiri DK. Epigenetics of dementia: understanding the disease as a transformation rather than a state. Lancet Neurol. 2016;15(7):760–774. doi: 10.1016/S1474-4422(16)00065-X.
    1. Itoh T, Satou T, Nishida S, Tsubaki M, Hashimoto S, Ito H. Improvement of cerebral function by anti-amyloid precursor protein antibody infusion after traumatic brain injury in rats. Mol Cell Biochem. 2009;324(1-2):191–199. doi: 10.1007/s11010-008-0013-1.
    1. Eriksson PS, Perfilieva E, Bjork-Eriksson T, Alborn AM, Nordborg C, Peterson DA, et al. Neurogenesis in the adult human hippocampus. Nat Med. 1998;4:1313–1317. doi: 10.1038/3305.
    1. Jin K, Minami M, Lan JQ, Mao XO, Batteur S, Simon RP, et al. Neurogenesis in dentate subgranular zone and rostral subventricular zone after focal cerebral ischemia in the rat. PNAS. 2001;98:4710–4715. doi: 10.1073/pnas.081011098.
    1. Chang EH, Adorjan I, Mundim MV, Sun B, Dizon ML, Szele FG. Traumatic brain injury activation of the adult subventricular zone Neurogenic niche. Front Neurosci. 2016;10:332.
    1. Vivar C, Potter MC, Choi J, Lee JY, Stringer TP, Callaway EM, et al. Monosynaptic inputs to new neurons in the dentate gyrus. Nat Commun. 2012;3:1107. doi: 10.1038/ncomms2101.
    1. Vivar C, van Praag H. Functional circuits of new neurons in the dentate gyrus. Front Neural Circuits. 2013;7:15. doi: 10.3389/fncir.2013.00015.
    1. Luo Y, Kuo CC, Shen H, Chou J, Greig NH, Hoffer BJ, et al. Delayed treatment with a p53 inhibitor enhances recovery in stroke brain. Ann Neurol. 2009;65:520–530. doi: 10.1002/ana.21592.
    1. Sun D. Endogenous neurogenic cell response in the mature mammalian brain following traumatic injury. Exp Neurol. 2016;275(Pt 3):405–410. doi: 10.1016/j.expneurol.2015.04.017.
    1. Loane DJ, Kumar A. Microglia in the TBI brain: the good, the bad, and the dysregulated. Exp Neurol. 2016;275(Pt 3):316–327. doi: 10.1016/j.expneurol.2015.08.018.
    1. Marutle A, Ohmitsu M, Nilbratt M, Greig NH, Nordberg A, Sugaya K. Modulation of human neural stem cell differentiation in Alzheimer (APP23) transgenic mice by phenserine. PNAS. 2007;104:12506–12511. doi: 10.1073/pnas.0705346104.
    1. Lilja AM, Röjdner J, Mustafiz T, Thomé CM, Storelli E, Gonzalez D, et al. Modulation of amyloid-β enhances hippocampal neurogenesis in adult Tg2576 mice. PLoS One. 2013;8(3):e58752. doi: 10.1371/journal.pone.0058752.
    1. Graham DI, Ford I, Adams JH, et al. Ischaemic brain damage is still common in fatal non-missile head injury. J Neurol Neurosurg Psychiatry. 1989;52:346–350. doi: 10.1136/jnnp.52.3.346.
    1. Miller JD. Head injury and brain ischaemia-implications for therapy. Br J Anaesth. 1985;57:120–130. doi: 10.1093/bja/57.1.120.
    1. Veenith TV, Carter EL, Geeraerts T, Grossac J, Newcombe VF, Outtrim J, et al. Pathophysiologic mechanisms of cerebral ischemia and diffusion hypoxia in traumatic brain injury. JAMA Neurol. 2016;73:542–550. doi: 10.1001/jamaneurol.2016.0091.
    1. Wang Y, Greig NH, Yu QS, Mattson MP. Presenilin-1 mutation impairs cholinergic modulation of synaptic plasticity and suppresses NMDA currents in hippocampus slices. Neurobiol Aging. 2009;30:1061–1068. doi: 10.1016/j.neurobiolaging.2007.10.009.
    1. Greig NH, Sambamurti K, Yu QS, Brossi A, Bruinsma GB, Lahiri DK. An overview of phenserine tartrate, a novel acetylcholinesterase inhibitor for the treatment of Alzheimer's disease. Curr Alzheimer Res. 2005;2:281–290. doi: 10.2174/1567205054367829.
    1. Greig NH, De Micheli E, Holloway HW, Yu QS, Utsuki T, Perry TA, et al. The experimental Alzheimer drug phenserine: preclinical pharmacokinetics and pharmacodynamics. Acta Neurol Scand Suppl. 2000;176:74–84. doi: 10.1034/j.1600-0404.2000.00311.x.
    1. Deselms H, Maggio N, Rubovitch V, Chapman J, Schreiber S, Tweedie D, et al. Novel pharmaceutical treatments for minimal traumatic brain injury and evaluation of animal models and methodologies supporting their development. J Neurosci Methods. 2016;272:69–76. doi: 10.1016/j.jneumeth.2016.02.002.
    1. Baratz R, Tweedie D, Wang JY, Rubovitch V, Luo W, Hoffer BJ, et al. Transiently lowering tumor necrosis factor-α synthesis ameliorates neuronal cell loss and cognitive impairments induced by minimal traumatic brain injury in mice. J Neuroinflammation. 2015;12:45. doi: 10.1186/s12974-015-0237-4.
    1. Kadir A, Andreasen N, Almkvist O, Wall A, Forsberg A, Engler H, et al. Effect of phenserine treatment on brain functional activity and amyloid in Alzheimer's disease. Ann Neurol. 2008;63:621–631. doi: 10.1002/ana.21345.

Source: PubMed

3
Iratkozz fel