Skip to main content
Top
Published in: Journal of Neuroinflammation 1/2015

Open Access 01-12-2015 | Research

Transiently lowering tumor necrosis factor-α synthesis ameliorates neuronal cell loss and cognitive impairments induced by minimal traumatic brain injury in mice

Authors: Renana Baratz, David Tweedie, Jia-Yi Wang, Vardit Rubovitch, Weiming Luo, Barry J Hoffer, Nigel H Greig, Chaim G Pick

Published in: Journal of Neuroinflammation | Issue 1/2015

Login to get access

Abstract

Background

The treatment of traumatic brain injury (TBI) represents an unmet medical need, as no effective pharmacological treatment currently exists. The development of such a treatment requires a fundamental understanding of the pathophysiological mechanisms that underpin the sequelae resulting from TBI, particularly the ensuing neuronal cell death and cognitive impairments. Tumor necrosis factor-alpha (TNF-α) is a cytokine that is a master regulator of systemic and neuroinflammatory processes. TNF-α levels are reported to become rapidly elevated post TBI and, potentially, can lead to secondary neuronal damage.

Methods

To elucidate the role of TNF-α in TBI, particularly as a drug target, the present study evaluated (i) time-dependent TNF-α levels and (ii) markers of apoptosis and gliosis within the brain and related these to behavioral measures of ‘well being’ and cognition in a mouse closed head 50 g weight drop mild TBI (mTBI) model in the presence and absence of post-treatment with an experimental TNF-α synthesis inhibitor, 3,6′-dithiothalidomide.

Results

mTBI elevated brain TNF-α levels, which peaked at 12 h post injury and returned to baseline by 18 h. This was accompanied by a neuronal loss and an increase in astrocyte number (evaluated by neuronal nuclei (NeuN) and glial fibrillary acidic protein (GFAP) immunostaining), as well as an elevation in the apoptotic death marker BH3-interacting domain death agonist (BID) at 72 h. Selective impairments in measures of cognition, evaluated by novel object recognition and passive avoidance paradigms - without changes in well being, were evident at 7 days after injury. A single systemic treatment with the TNF-α synthesis inhibitor 3,6′-dithiothalidomide 1 h post injury prevented the mTBI-induced TNF-α elevation and fully ameliorated the neuronal loss (NeuN), elevations in astrocyte number (GFAP) and BID, and cognitive impairments. Cognitive impairments evident at 7 days after injury were prevented by treatment as late as 12 h post mTBI but were not reversed when treatment was delayed until 18 h.

Conclusions

These results implicate that TNF-α in mTBI induced secondary brain damage and indicate that pharmacologically limiting the generation of TNF-α post mTBI may mitigate such damage, defining a time-dependent window of up to 12 h to achieve this reversal.
Literature
1.
go back to reference Hyder AA, Wunderlich CA, Puvanachandra P, Gururaj G, Kobusingye OC. The impact of traumatic brain injuries: a global perspective. NeuroRehabilitation. 2007;22:341–53.PubMed Hyder AA, Wunderlich CA, Puvanachandra P, Gururaj G, Kobusingye OC. The impact of traumatic brain injuries: a global perspective. NeuroRehabilitation. 2007;22:341–53.PubMed
2.
go back to reference Setnik L, Bazarian JJ. The characteristics of patients who do not seek medical treatment for traumatic brain injury. Brain Inj. 2007;21(1):1–9.CrossRefPubMed Setnik L, Bazarian JJ. The characteristics of patients who do not seek medical treatment for traumatic brain injury. Brain Inj. 2007;21(1):1–9.CrossRefPubMed
3.
go back to reference Faul M, Xu L, Wald M, Coronado VG. Traumatic Brain Injury in the United States: Emergency Department Visits, Hospitalizations and Deaths 2002–2006. Atlanta (GA): Centers for Disease Control and Prevention, National Center for Injury Prevention and Control; 2010. Faul M, Xu L, Wald M, Coronado VG. Traumatic Brain Injury in the United States: Emergency Department Visits, Hospitalizations and Deaths 2002–2006. Atlanta (GA): Centers for Disease Control and Prevention, National Center for Injury Prevention and Control; 2010.
4.
go back to reference McGarry LJ, Thompson D, Millham FH, Cowell L, Snyder PJ, Lenderking WR, et al. Outcomes and costs of acute treatment of traumatic brain injury. J Trauma. 2002;53(6):1152–9.CrossRefPubMed McGarry LJ, Thompson D, Millham FH, Cowell L, Snyder PJ, Lenderking WR, et al. Outcomes and costs of acute treatment of traumatic brain injury. J Trauma. 2002;53(6):1152–9.CrossRefPubMed
5.
go back to reference McAllister TW. Neurobehavioral sequelae of traumatic brain injury: evaluation and management. World Psychiatry. 2008;7(1):3–10.PubMedCentralPubMed McAllister TW. Neurobehavioral sequelae of traumatic brain injury: evaluation and management. World Psychiatry. 2008;7(1):3–10.PubMedCentralPubMed
6.
go back to reference Coronado VG, Xu L, Basavaraju SV, McGuire LC, Wald MM, Faul MD, et al. Centers for Disease Control and Prevention (CDC). Surveillance for traumatic brain injury-related deaths–United States, 1997–2007. MMWR Surveill Summ. 2011;60(5):1–32.PubMed Coronado VG, Xu L, Basavaraju SV, McGuire LC, Wald MM, Faul MD, et al. Centers for Disease Control and Prevention (CDC). Surveillance for traumatic brain injury-related deaths–United States, 1997–2007. MMWR Surveill Summ. 2011;60(5):1–32.PubMed
7.
go back to reference Yu S, Kaneko Y, Bae E, Stahl CE, Wang Y, van Loveren H, et al. Severity of controlled cortical impact traumatic brain injury in rats and mice dictates degree of behavioral deficits. Brain Res. 2009;1287:157–63.CrossRefPubMed Yu S, Kaneko Y, Bae E, Stahl CE, Wang Y, van Loveren H, et al. Severity of controlled cortical impact traumatic brain injury in rats and mice dictates degree of behavioral deficits. Brain Res. 2009;1287:157–63.CrossRefPubMed
8.
go back to reference Iverson GL. Outcome from mild traumatic brain injury. Curr Opinion Psychiatry. 2005;18:301–7.CrossRef Iverson GL. Outcome from mild traumatic brain injury. Curr Opinion Psychiatry. 2005;18:301–7.CrossRef
9.
go back to reference Schreiber S, Barkai G, Gur-Hartman T, Peles E, Tov N, Dolberg OT, et al. Long-lasting sleep patterns of adult patients with minor traumatic brain injury (mTBI) and non-mTBI subjects. Sleep Med. 2008;9:481–7.CrossRefPubMed Schreiber S, Barkai G, Gur-Hartman T, Peles E, Tov N, Dolberg OT, et al. Long-lasting sleep patterns of adult patients with minor traumatic brain injury (mTBI) and non-mTBI subjects. Sleep Med. 2008;9:481–7.CrossRefPubMed
10.
go back to reference Mortimer JA, van Duijn CM, Chandra V, Fratiglioni L, Graves AB, Heyman A, et al. Head trauma as a risk factor for Alzheimer’s disease: a collaborative re-analysis of case–control studies, EURODEM Risk Factors Research Group. Int J Epidemiol. 1991;20 Suppl 2:S28–35.CrossRefPubMed Mortimer JA, van Duijn CM, Chandra V, Fratiglioni L, Graves AB, Heyman A, et al. Head trauma as a risk factor for Alzheimer’s disease: a collaborative re-analysis of case–control studies, EURODEM Risk Factors Research Group. Int J Epidemiol. 1991;20 Suppl 2:S28–35.CrossRefPubMed
11.
go back to reference Mayeux R, Ottman R, Maestre G, Ngai C, Tang MX, Ginsberg H, et al. Synergistic effects of traumatic head-injury and apolipoprotein-epsilon-4 in patients with Alzheimers-disease. Neurology. 1995;45:555–7.CrossRefPubMed Mayeux R, Ottman R, Maestre G, Ngai C, Tang MX, Ginsberg H, et al. Synergistic effects of traumatic head-injury and apolipoprotein-epsilon-4 in patients with Alzheimers-disease. Neurology. 1995;45:555–7.CrossRefPubMed
12.
go back to reference 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.CrossRefPubMedCentralPubMed 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.CrossRefPubMedCentralPubMed
13.
go back to reference Goldstein LE, Fisher AM, Tagge CA, Zhang XL, Velisek L, Sullivan JA, et al. Chronic traumatic encephalopathy in blast-exposed military veterans and a blast neurotrauma mouse model. Sci Transl Med. 2012;4(134):134ra60.CrossRefPubMedCentralPubMed Goldstein LE, Fisher AM, Tagge CA, Zhang XL, Velisek L, Sullivan JA, et al. Chronic traumatic encephalopathy in blast-exposed military veterans and a blast neurotrauma mouse model. Sci Transl Med. 2012;4(134):134ra60.CrossRefPubMedCentralPubMed
14.
go back to reference 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):312–9.CrossRefPubMed 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):312–9.CrossRefPubMed
15.
go back to reference 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 [Epub ahead of print]. 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 [Epub ahead of print].
16.
go back to reference Kumar A, Stoica BA, Sabirzhanov B, Burns MP, Faden AI, Loane DJ. Traumatic brain injury in aged animals increases lesion size and chronically alters microglial/macrophage classical and alternative activation states. Neurobiol Aging. 2013;34(5):1397–411.CrossRefPubMedCentralPubMed Kumar A, Stoica BA, Sabirzhanov B, Burns MP, Faden AI, Loane DJ. Traumatic brain injury in aged animals increases lesion size and chronically alters microglial/macrophage classical and alternative activation states. Neurobiol Aging. 2013;34(5):1397–411.CrossRefPubMedCentralPubMed
17.
go back to reference Loane DJ, Faden AI. Neuroprotection for traumatic brain injury: translational challenges and emerging therapeutic strategies. Trends Pharmacol Sci. 2010;31(12):596–604.CrossRefPubMedCentralPubMed Loane DJ, Faden AI. Neuroprotection for traumatic brain injury: translational challenges and emerging therapeutic strategies. Trends Pharmacol Sci. 2010;31(12):596–604.CrossRefPubMedCentralPubMed
18.
go back to reference Diaz-Arrastia R, Kochanek PM, Bergold P, Kenney K, Marx CE, Grimes CJ, 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.CrossRefPubMedCentralPubMed Diaz-Arrastia R, Kochanek PM, Bergold P, Kenney K, Marx CE, Grimes CJ, 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.CrossRefPubMedCentralPubMed
19.
go back to reference Finnie JW, Blumbergs PC, Manavis J, Vink R. Pattern of cerebrospinal immediate early gene c-fos expression in an ovine model of non-accidental head injury. J Clin Neurosci. 2013;20:1759–61.CrossRefPubMed Finnie JW, Blumbergs PC, Manavis J, Vink R. Pattern of cerebrospinal immediate early gene c-fos expression in an ovine model of non-accidental head injury. J Clin Neurosci. 2013;20:1759–61.CrossRefPubMed
20.
go back to reference Greig NH, Tweedie D, Rachmany L, Li Y, Rubovitch V, Schreiber S, 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):S62–75.CrossRefPubMedCentralPubMed Greig NH, Tweedie D, Rachmany L, Li Y, Rubovitch V, Schreiber S, 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):S62–75.CrossRefPubMedCentralPubMed
21.
go back to reference Barkhoudarian G, Hovda DA, Giza C. The molecular pathophysiology of concussive brain injury. Clin Sports Med. 2011;30:33–48.CrossRefPubMed Barkhoudarian G, Hovda DA, Giza C. The molecular pathophysiology of concussive brain injury. Clin Sports Med. 2011;30:33–48.CrossRefPubMed
22.
24.
go back to reference Tweedie D, Milman A, Holloway HW, Li YZ, Harvey BK, Shen H, et al. Apoptotic and behavioral sequelae of mild brain trauma in mice. J Neurosci Res. 2007;85:805–15.CrossRefPubMed Tweedie D, Milman A, Holloway HW, Li YZ, Harvey BK, Shen H, et al. Apoptotic and behavioral sequelae of mild brain trauma in mice. J Neurosci Res. 2007;85:805–15.CrossRefPubMed
25.
go back to reference Stoica BA, Byrnes KR, Faden AI. Cell cycle activation and CNS injury. Neurotox Res. 2009;16:221–37.CrossRefPubMed Stoica BA, Byrnes KR, Faden AI. Cell cycle activation and CNS injury. Neurotox Res. 2009;16:221–37.CrossRefPubMed
26.
go back to reference Morganti-Kossmann MC, Rancan M, Stahel PF, Kossmann T. Inflammatory response in acute traumatic brain injury: a double-edged sword. Curr Opinion Crit Care. 2002;8:101–5.CrossRef Morganti-Kossmann MC, Rancan M, Stahel PF, Kossmann T. Inflammatory response in acute traumatic brain injury: a double-edged sword. Curr Opinion Crit Care. 2002;8:101–5.CrossRef
27.
go back to reference Stoll G, Jander S, Schroeter M. Detrimental and beneficial effects of injury-induced inflammation and cytokine expression in the nervous system. Adv Exp Med Biol. 2002;513:87–113.PubMed Stoll G, Jander S, Schroeter M. Detrimental and beneficial effects of injury-induced inflammation and cytokine expression in the nervous system. Adv Exp Med Biol. 2002;513:87–113.PubMed
28.
go back to reference Schmidt OI, Heyde CE, Ertel W, Stahel PF. Closed head injury - an inflammatory disease? Brain Res Rev. 2005;48:388–99.CrossRefPubMed Schmidt OI, Heyde CE, Ertel W, Stahel PF. Closed head injury - an inflammatory disease? Brain Res Rev. 2005;48:388–99.CrossRefPubMed
29.
go back to reference McCoy MK, Tansey MG. TNF signaling inhibition in the CNS: implications for normal brain function and neurodegenerative disease. J Neuroinflammation. 2008;5:45.CrossRefPubMedCentralPubMed McCoy MK, Tansey MG. TNF signaling inhibition in the CNS: implications for normal brain function and neurodegenerative disease. J Neuroinflammation. 2008;5:45.CrossRefPubMedCentralPubMed
30.
go back to reference 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(3):391–403.CrossRefPubMed 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(3):391–403.CrossRefPubMed
31.
go back to reference Clark IA, Alleva LM, Vissel B. The roles of TNF in brain dysfunction and disease. Pharmacol Ther. 2010;128(3):519–48.CrossRefPubMed Clark IA, Alleva LM, Vissel B. The roles of TNF in brain dysfunction and disease. Pharmacol Ther. 2010;128(3):519–48.CrossRefPubMed
32.
go back to reference Frugier T, Morganti-Kossmann MC, O’Reilly D, McLean CA. In situ detection of inflammatory mediators in post mortem human brain tissue after traumatic injury. J Neurotrauma. 2010;27:497–507.CrossRefPubMed Frugier T, Morganti-Kossmann MC, O’Reilly D, McLean CA. In situ detection of inflammatory mediators in post mortem human brain tissue after traumatic injury. J Neurotrauma. 2010;27:497–507.CrossRefPubMed
33.
go back to reference Shohami E, Gallily R, Mechoulam R, Bass R, Ben-Hur T. Cytokine production in the brain following closed head injury: dexanabinol (HU-211) is a novel TNF-α inhibitor and an effective neuroprotectant. J Neuroimmunol. 1997;72:169–77.CrossRefPubMed Shohami E, Gallily R, Mechoulam R, Bass R, Ben-Hur T. Cytokine production in the brain following closed head injury: dexanabinol (HU-211) is a novel TNF-α inhibitor and an effective neuroprotectant. J Neuroimmunol. 1997;72:169–77.CrossRefPubMed
34.
go back to reference Lu J, Goh SJ, Tng PY, Deng YY, Ling EA, Moochhala S. Systemic inflammatory response following acute traumatic brain injury. Front Biosci. 2009;14:3795–813.CrossRef Lu J, Goh SJ, Tng PY, Deng YY, Ling EA, Moochhala S. Systemic inflammatory response following acute traumatic brain injury. Front Biosci. 2009;14:3795–813.CrossRef
35.
go back to reference Yang J, You Z, Kim HH, Hwang SK, Khuman J, Guo S, et al. Genetic analysis of the role of tumor necrosis factor receptors in functional outcome after traumatic brain injury in mice. J Neurotrauma. 2010;27:1037–46.CrossRefPubMedCentralPubMed Yang J, You Z, Kim HH, Hwang SK, Khuman J, Guo S, et al. Genetic analysis of the role of tumor necrosis factor receptors in functional outcome after traumatic brain injury in mice. J Neurotrauma. 2010;27:1037–46.CrossRefPubMedCentralPubMed
36.
go back to reference Tuttolomondo A, Pecoraro R, Pinto A. Studies of selective TNF inhibitors in the treatment of brain injury from stroke and trauma: a review of the evidence to date. Drug Des Devel Ther. 2014;8:2221–38.CrossRefPubMedCentralPubMed Tuttolomondo A, Pecoraro R, Pinto A. Studies of selective TNF inhibitors in the treatment of brain injury from stroke and trauma: a review of the evidence to date. Drug Des Devel Ther. 2014;8:2221–38.CrossRefPubMedCentralPubMed
38.
go back to reference Santello M, Volterra A. TNFα in synaptic function: switching gears. Trends Neurosci. 2012;35(10):638–47.CrossRefPubMed Santello M, Volterra A. TNFα in synaptic function: switching gears. Trends Neurosci. 2012;35(10):638–47.CrossRefPubMed
39.
go back to reference Bermpohl D, You Z, Lo EH, Kim HH, Whalen MJ. TNF alpha and Fas mediate tissue damage and functional outcome after traumatic brain injury in mice. J Cerebral Blood Flow Metabol. 2007;27:1806–18.CrossRef Bermpohl D, You Z, Lo EH, Kim HH, Whalen MJ. TNF alpha and Fas mediate tissue damage and functional outcome after traumatic brain injury in mice. J Cerebral Blood Flow Metabol. 2007;27:1806–18.CrossRef
40.
go back to reference Longhi L, Ortolano F, Zanier ER, Perego C, Stocchetti N, De Simoni MG. Effect of traumatic brain injury on cognitive function in mice lacking p55 and p75 tumor necrosis factor receptors. Acta Neurochir Suppl. 2008;102:409–13.CrossRefPubMed Longhi L, Ortolano F, Zanier ER, Perego C, Stocchetti N, De Simoni MG. Effect of traumatic brain injury on cognitive function in mice lacking p55 and p75 tumor necrosis factor receptors. Acta Neurochir Suppl. 2008;102:409–13.CrossRefPubMed
41.
go back to reference Longhi L, Perego C, Ortolano F, Aresi S, Fumagalli S, Zanier ER, et al. Tumor necrosis factor in traumatic brain injury: effects of genetic deletion of p55 or p75 receptor. J Cereb Blood Flow Metab. 2013;33(8):1182–9.CrossRefPubMedCentralPubMed Longhi L, Perego C, Ortolano F, Aresi S, Fumagalli S, Zanier ER, et al. Tumor necrosis factor in traumatic brain injury: effects of genetic deletion of p55 or p75 receptor. J Cereb Blood Flow Metab. 2013;33(8):1182–9.CrossRefPubMedCentralPubMed
42.
go back to reference Scherbel U, Raghupathi R, Nakamura M, Saatman KE, Trojanowski JQ, Neugebauer E, et al. Differential acute and chronic responses of tumor necrosis factor-deficient mice to experimental brain injury. Proc Natl Acad Sci U S A. 1999;96:8721–6.CrossRefPubMedCentralPubMed Scherbel U, Raghupathi R, Nakamura M, Saatman KE, Trojanowski JQ, Neugebauer E, et al. Differential acute and chronic responses of tumor necrosis factor-deficient mice to experimental brain injury. Proc Natl Acad Sci U S A. 1999;96:8721–6.CrossRefPubMedCentralPubMed
43.
go back to reference Zhu X, Giordano T, Yu QS, Holloway HW, Perry T, Lahiri DK, et al. Thiothalidomides: novel isosteric analogues of thalidomide with enhanced TNF-alpha inhibitory activity. J Med Chem. 2003;46:5222–9.CrossRefPubMed Zhu X, Giordano T, Yu QS, Holloway HW, Perry T, Lahiri DK, et al. Thiothalidomides: novel isosteric analogues of thalidomide with enhanced TNF-alpha inhibitory activity. J Med Chem. 2003;46:5222–9.CrossRefPubMed
44.
go back to reference Baratz R, Tweedie D, Rubovitch V, Luo W, Yoon JS, Hoffer BJ, et al. Tumor necrosis factor-alpha synthesis inhibitor, 3,6′-dithiothalidomide, reverses behavioral impairments induced by minimal traumatic brain injury in mice. J Neurochem. 2011;118:1032–42.CrossRefPubMedCentralPubMed Baratz R, Tweedie D, Rubovitch V, Luo W, Yoon JS, Hoffer BJ, et al. Tumor necrosis factor-alpha synthesis inhibitor, 3,6′-dithiothalidomide, reverses behavioral impairments induced by minimal traumatic brain injury in mice. J Neurochem. 2011;118:1032–42.CrossRefPubMedCentralPubMed
45.
go back to reference 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;118:949–55.CrossRefPubMed 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;118:949–55.CrossRefPubMed
46.
go back to reference Milman A, Rosenberg A, Weizman R, Pick CG. Mild traumatic brain injury induces persistent cognitive deficits and behavioral disturbances in mice. J Neurotrauma. 2005;22:1003–10.CrossRefPubMed Milman A, Rosenberg A, Weizman R, Pick CG. Mild traumatic brain injury induces persistent cognitive deficits and behavioral disturbances in mice. J Neurotrauma. 2005;22:1003–10.CrossRefPubMed
47.
go back to reference Hogg S. A review of the validity and variability of the elevated plus-maze as an animal model of anxiety. Pharmacol Biochem Behav. 1996;54:21–30.CrossRefPubMed Hogg S. A review of the validity and variability of the elevated plus-maze as an animal model of anxiety. Pharmacol Biochem Behav. 1996;54:21–30.CrossRefPubMed
48.
go back to reference Dellu F, Mayo W, Cherkaoui J, Lemoal M, Simon H. A 2-trial memory task with automated recording - study in young and aged rats. Brain Res. 1992;588:132–9.CrossRefPubMed Dellu F, Mayo W, Cherkaoui J, Lemoal M, Simon H. A 2-trial memory task with automated recording - study in young and aged rats. Brain Res. 1992;588:132–9.CrossRefPubMed
49.
go back to reference Dix SL, Aggleton JP. Extending the spontaneous preference test of recognition: evidence of object-location and object-context recognition. Behav Brain Res. 1999;99:191–200.CrossRefPubMed Dix SL, Aggleton JP. Extending the spontaneous preference test of recognition: evidence of object-location and object-context recognition. Behav Brain Res. 1999;99:191–200.CrossRefPubMed
50.
go back to reference Meisser C. Object recognition in mice: improvement of memory by glucose. Neurobiol Learning Memory. 1997;67:172–5.CrossRef Meisser C. Object recognition in mice: improvement of memory by glucose. Neurobiol Learning Memory. 1997;67:172–5.CrossRef
51.
go back to reference Edut S, Rubovitch V, Schreiber S, Pick CG. The intriguing effects of ecstasy (MDMA) on cognitive function in mice subjected to a minimal traumatic brain injury (mTBI). Psychopharmacol. 2011;214:877–89.CrossRef Edut S, Rubovitch V, Schreiber S, Pick CG. The intriguing effects of ecstasy (MDMA) on cognitive function in mice subjected to a minimal traumatic brain injury (mTBI). Psychopharmacol. 2011;214:877–89.CrossRef
52.
go back to reference Amaral D, Scharfman H, Lavenex P. The dentate gyrus: fundamental neuroanatomical organization (dentate gyrus for dummies). Prog Brain Res. 2007;163:3–22. 788–790.CrossRefPubMedCentralPubMed Amaral D, Scharfman H, Lavenex P. The dentate gyrus: fundamental neuroanatomical organization (dentate gyrus for dummies). Prog Brain Res. 2007;163:3–22. 788–790.CrossRefPubMedCentralPubMed
53.
go back to reference Saab BJ, Georgiou J, Nath A, Lee FJ, Wang M, Michalon A, et al. NCS-1 in the dentate gyrus promotes exploration, synaptic plasticity, and rapid acquisition of spatial memory. Neuron. 2009;63(5):643–56.CrossRefPubMed Saab BJ, Georgiou J, Nath A, Lee FJ, Wang M, Michalon A, et al. NCS-1 in the dentate gyrus promotes exploration, synaptic plasticity, and rapid acquisition of spatial memory. Neuron. 2009;63(5):643–56.CrossRefPubMed
54.
55.
go back to reference Tobinick E, Kim NM, Reyzin G, Rodriguez-Romanacce H, DePuy V. Selective TNF inhibition for chronic stroke and traumatic brain injury: an observational study involving 629 consecutive patients treated with perispinal etanercept. CNS Drugs. 2012;26(12):1051–70.CrossRefPubMed Tobinick E, Kim NM, Reyzin G, Rodriguez-Romanacce H, DePuy V. Selective TNF inhibition for chronic stroke and traumatic brain injury: an observational study involving 629 consecutive patients treated with perispinal etanercept. CNS Drugs. 2012;26(12):1051–70.CrossRefPubMed
56.
go back to reference Tuttolomondo A, Di Raimondo D, di Sciacca R, Pinto A, Licata G. Inflammatory cytokines in acute ischemic stroke. Curr Pharm Des. 2008;14:3574–89.CrossRefPubMed Tuttolomondo A, Di Raimondo D, di Sciacca R, Pinto A, Licata G. Inflammatory cytokines in acute ischemic stroke. Curr Pharm Des. 2008;14:3574–89.CrossRefPubMed
57.
go back to reference 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;239:170–82.CrossRefPubMedCentralPubMed 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;239:170–82.CrossRefPubMedCentralPubMed
58.
go back to reference Sharp DJ, Scott G, Leech R. Network dysfunction after traumatic brain injury. Nat Rev Neurol. 2014;10(3):156–66.CrossRefPubMed Sharp DJ, Scott G, Leech R. Network dysfunction after traumatic brain injury. Nat Rev Neurol. 2014;10(3):156–66.CrossRefPubMed
59.
go back to reference Ryu WH, Feinstein A, Colantonio A, Streiner DL, Dawson DR. Early identification and incidence of mild TBI in Ontario. Can J Neurol Sci. 2009;36(4):429–35.CrossRefPubMed Ryu WH, Feinstein A, Colantonio A, Streiner DL, Dawson DR. Early identification and incidence of mild TBI in Ontario. Can J Neurol Sci. 2009;36(4):429–35.CrossRefPubMed
60.
go back to reference Moppett IK. Traumatic brain injury: assessment, resuscitation and early management. Br J Anaesth. 2007;99:18–31.CrossRefPubMed Moppett IK. Traumatic brain injury: assessment, resuscitation and early management. Br J Anaesth. 2007;99:18–31.CrossRefPubMed
61.
go back to reference Wajant H, Pfizenmaier K, Scheurich P. Tumor necrosis factor signaling. Cell Death Differ. 2003;10(1):45–65.CrossRefPubMed Wajant H, Pfizenmaier K, Scheurich P. Tumor necrosis factor signaling. Cell Death Differ. 2003;10(1):45–65.CrossRefPubMed
62.
go back to reference Grell M, Wajant H, Zimmermann G, Scheurich P. The type 1 receptor (CD120a) is the high-affinity receptor for soluble tumor necrosis factor. Proc Natl Acad Sci U S A. 1998;95(2):570–5.CrossRefPubMedCentralPubMed Grell M, Wajant H, Zimmermann G, Scheurich P. The type 1 receptor (CD120a) is the high-affinity receptor for soluble tumor necrosis factor. Proc Natl Acad Sci U S A. 1998;95(2):570–5.CrossRefPubMedCentralPubMed
63.
go back to reference Fontaine V, Mohand-Said S, Hanoteau N, Fuchs C, Pfizenmaier K, Eisel U. Neurodegenerative and neuroprotective effects of tumor necrosis factor (TNF) in retinal ischemia: opposite roles of TNF receptor 1 and TNF receptor 2. J Neurosci. 2002;22(7):RC216.PubMed Fontaine V, Mohand-Said S, Hanoteau N, Fuchs C, Pfizenmaier K, Eisel U. Neurodegenerative and neuroprotective effects of tumor necrosis factor (TNF) in retinal ischemia: opposite roles of TNF receptor 1 and TNF receptor 2. J Neurosci. 2002;22(7):RC216.PubMed
64.
go back to reference Yang L, Lindholm K, Konishi Y, Li R, Shen Y. Target depletion of distinct tumor necrosis factor receptor subtypes reveals hippocampal neuron death and survival through different signal transduction pathways. J Neurosci. 2002;22(8):3025–32.PubMed Yang L, Lindholm K, Konishi Y, Li R, Shen Y. Target depletion of distinct tumor necrosis factor receptor subtypes reveals hippocampal neuron death and survival through different signal transduction pathways. J Neurosci. 2002;22(8):3025–32.PubMed
65.
go back to reference Marchetti L, Klein M, Schlett K, Pfizenmaier K, Eisel UL. Tumor necrosis factor (TNF)-mediated neuroprotection against glutamate-induced excitotoxicity is enhanced by N-methyl-D-aspartate receptor activation. Essential role of a TNF receptor 2-mediated phosphatidylinositol 3-kinase-dependent NF-kappa B pathway. J Biol Chem. 2004;279(31):32869–81.CrossRefPubMed Marchetti L, Klein M, Schlett K, Pfizenmaier K, Eisel UL. Tumor necrosis factor (TNF)-mediated neuroprotection against glutamate-induced excitotoxicity is enhanced by N-methyl-D-aspartate receptor activation. Essential role of a TNF receptor 2-mediated phosphatidylinositol 3-kinase-dependent NF-kappa B pathway. J Biol Chem. 2004;279(31):32869–81.CrossRefPubMed
66.
go back to reference Grell M, Douni E, Wajant H, Löhden M, Clauss M, Maxeiner B, et al. The transmembrane form of tumor necrosis factor is the prime activating ligand of the 80 kDa tumor necrosis factor receptor. Cell. 1995;83(5):793–802.CrossRefPubMed Grell M, Douni E, Wajant H, Löhden M, Clauss M, Maxeiner B, et al. The transmembrane form of tumor necrosis factor is the prime activating ligand of the 80 kDa tumor necrosis factor receptor. Cell. 1995;83(5):793–802.CrossRefPubMed
67.
go back to reference Grell M. Tumor necrosis factor (TNF) receptors in cellular signaling of soluble and membrane-expressed TNF. J Inflamm. 1995–1996;47(1–2):8–17 Grell M. Tumor necrosis factor (TNF) receptors in cellular signaling of soluble and membrane-expressed TNF. J Inflamm. 1995–1996;47(1–2):8–17
68.
69.
go back to reference Tweedie D, Sambamurti K, Greig NH. TNF-α inhibition as a treatment strategy for neurodegenerative disorders: new drug candidates and targets. Curr Alzheimer Res. 2007;4:378–85.CrossRefPubMed Tweedie D, Sambamurti K, Greig NH. TNF-α inhibition as a treatment strategy for neurodegenerative disorders: new drug candidates and targets. Curr Alzheimer Res. 2007;4:378–85.CrossRefPubMed
70.
go back to reference Ponsford J, Cameron P, Fitzgerald M, Grant M, Mikocka-Walus A. Long-term outcomes after uncomplicated mild traumatic brain injury: a comparison with trauma controls. J Neurotrauma. 2011;28:937–46.CrossRefPubMed Ponsford J, Cameron P, Fitzgerald M, Grant M, Mikocka-Walus A. Long-term outcomes after uncomplicated mild traumatic brain injury: a comparison with trauma controls. J Neurotrauma. 2011;28:937–46.CrossRefPubMed
71.
go back to reference Ghajar J, Ivry RB. Cognitive and Neurobiological Research Consortium. The predictive brain state: timing deficiency in traumatic brain injury? Neurorehabil Neural Repair. 2008;22(3):217–27.CrossRefPubMedCentralPubMed Ghajar J, Ivry RB. Cognitive and Neurobiological Research Consortium. The predictive brain state: timing deficiency in traumatic brain injury? Neurorehabil Neural Repair. 2008;22(3):217–27.CrossRefPubMedCentralPubMed
72.
go back to reference Israelsson C, Bengtsson H, Kylberg A, Kullander K, Lewén A, Hillered L, et al. Distinct cellular patterns of upregulated chemokine expression supporting a prominent inflammatory role in traumatic brain injury. J Neurotrauma. 2008;25(8):959–74.CrossRefPubMed Israelsson C, Bengtsson H, Kylberg A, Kullander K, Lewén A, Hillered L, et al. Distinct cellular patterns of upregulated chemokine expression supporting a prominent inflammatory role in traumatic brain injury. J Neurotrauma. 2008;25(8):959–74.CrossRefPubMed
73.
go back to reference Israelsson C, Wang Y, Kylberg A, Pick CG, Hoffer BJ, Ebendal T. Closed head injury in a mouse model results in molecular changes indicating inflammatory responses. J Neurotrauma. 2009;26(8):1307–14.CrossRefPubMedCentralPubMed Israelsson C, Wang Y, Kylberg A, Pick CG, Hoffer BJ, Ebendal T. Closed head injury in a mouse model results in molecular changes indicating inflammatory responses. J Neurotrauma. 2009;26(8):1307–14.CrossRefPubMedCentralPubMed
74.
go back to reference Taupin V, Toulmond S, Serrano A, Benavides J, Zavala F. Increase in IL-6, IL-1 and TNF levels in rat brain following traumatic lesion: influence of pre- and post-traumatic treatment with Ro5 4864, a peripheral-type (p site) benzodiazepine ligand. J Neuroimmunol. 1993;42:177–85.CrossRefPubMed Taupin V, Toulmond S, Serrano A, Benavides J, Zavala F. Increase in IL-6, IL-1 and TNF levels in rat brain following traumatic lesion: influence of pre- and post-traumatic treatment with Ro5 4864, a peripheral-type (p site) benzodiazepine ligand. J Neuroimmunol. 1993;42:177–85.CrossRefPubMed
75.
go back to reference Shohami E, Novikov M, Bass R, Yamin A, Gallily R. Closed head injury triggers early production of TNF-α and IL-6 by brain tissue. J Cereb Blood Flow Metab. 1994;14:615–9.CrossRefPubMed Shohami E, Novikov M, Bass R, Yamin A, Gallily R. Closed head injury triggers early production of TNF-α and IL-6 by brain tissue. J Cereb Blood Flow Metab. 1994;14:615–9.CrossRefPubMed
76.
go back to reference Gourin CG, Shackford SR. Production of tumor necrosis factor-alpha and interleukin-1beta by human cerebral microvascular endothelium after percussive trauma. J Trauma. 1997;42(6):1101–7.CrossRefPubMed Gourin CG, Shackford SR. Production of tumor necrosis factor-alpha and interleukin-1beta by human cerebral microvascular endothelium after percussive trauma. J Trauma. 1997;42(6):1101–7.CrossRefPubMed
77.
go back to reference Knoblach SM, Fan L, Faden AI. Early neuronal expression of tumor necrosis factor-alpha after experimental brain injury contributes to neurological impairment. J Neuroimmunol. 1999;95(1–2):115–25.CrossRefPubMed Knoblach SM, Fan L, Faden AI. Early neuronal expression of tumor necrosis factor-alpha after experimental brain injury contributes to neurological impairment. J Neuroimmunol. 1999;95(1–2):115–25.CrossRefPubMed
78.
go back to reference Ross SA, Halliday MI, Campbell GC, Byrnes DP, Rowlands BJ. The presence of tumour necrosis factor in CSF and plasma after severe head injury. Br J Neurosurg. 1994;8(4):419–25.CrossRefPubMed Ross SA, Halliday MI, Campbell GC, Byrnes DP, Rowlands BJ. The presence of tumour necrosis factor in CSF and plasma after severe head injury. Br J Neurosurg. 1994;8(4):419–25.CrossRefPubMed
79.
go back to reference Tweedie D, Ferguson RA, Fishman K, Frankola KA, Van Praag H, Holloway HW, et al. Tumor necrosis factor-α synthesis inhibitor 3,6′-dithiothalidomide attenuates markers of inflammation. Alzheimer pathology and behavioral deficits in animal models of neuroinflammation and Alzheimer’s disease. J Neuroinflammation. 2012;9:106.CrossRefPubMedCentralPubMed Tweedie D, Ferguson RA, Fishman K, Frankola KA, Van Praag H, Holloway HW, et al. Tumor necrosis factor-α synthesis inhibitor 3,6′-dithiothalidomide attenuates markers of inflammation. Alzheimer pathology and behavioral deficits in animal models of neuroinflammation and Alzheimer’s disease. J Neuroinflammation. 2012;9:106.CrossRefPubMedCentralPubMed
80.
go back to reference Gabbita SP, Srivastava MK, Eslami P, Johnson MF, Kobritz NK, Tweedie D, et al. Early intervention with a small molecule inhibitor for tumor necrosis factor-α prevents cognitive deficits in a triple transgenic mouse model of Alzheimer’s disease. J Neuroinflammation. 2012;9:99.CrossRefPubMedCentralPubMed Gabbita SP, Srivastava MK, Eslami P, Johnson MF, Kobritz NK, Tweedie D, et al. Early intervention with a small molecule inhibitor for tumor necrosis factor-α prevents cognitive deficits in a triple transgenic mouse model of Alzheimer’s disease. J Neuroinflammation. 2012;9:99.CrossRefPubMedCentralPubMed
81.
go back to reference Yoon JS, Lee JH, Tweedie D, Mughal MR, Chigurupati S, Greig NH, et al. 3,6′-Dithiothalidomide improves experimental stroke outcome by suppressing neuroinflammation. J Neurosci Res. 2013;91(5):671–80.CrossRefPubMed Yoon JS, Lee JH, Tweedie D, Mughal MR, Chigurupati S, Greig NH, et al. 3,6′-Dithiothalidomide improves experimental stroke outcome by suppressing neuroinflammation. J Neurosci Res. 2013;91(5):671–80.CrossRefPubMed
82.
go back to reference Tashlykov V, Katz Y, Volkov A, Gazit V, Schreiber S, Zohar O, et al. Minimal traumatic brain injury induce apoptotic cell death in mice. J Mol Neurosci. 2009;37(1):16–24.CrossRefPubMed Tashlykov V, Katz Y, Volkov A, Gazit V, Schreiber S, Zohar O, et al. Minimal traumatic brain injury induce apoptotic cell death in mice. J Mol Neurosci. 2009;37(1):16–24.CrossRefPubMed
83.
go back to reference Bussey TJ, Padain TL, Skillings EA, Winters BD, Morton AJ, Saksida LM. The touchscreen cognitive testing method for rodents: how to get the best out of your rat. Learn Mem. 2008;2008(15):516–23.CrossRef Bussey TJ, Padain TL, Skillings EA, Winters BD, Morton AJ, Saksida LM. The touchscreen cognitive testing method for rodents: how to get the best out of your rat. Learn Mem. 2008;2008(15):516–23.CrossRef
84.
go back to reference Tashlykov V, Katz Y, Gazit V, Zohar O, Schreiber S, Pick CG. Apoptotic changes in the cortex and hippocampus following minimal brain trauma in mice. Brain Res. 2007;1130(1):197–205.CrossRefPubMed Tashlykov V, Katz Y, Gazit V, Zohar O, Schreiber S, Pick CG. Apoptotic changes in the cortex and hippocampus following minimal brain trauma in mice. Brain Res. 2007;1130(1):197–205.CrossRefPubMed
85.
go back to reference Wakade C, Sukumari-Ramesh S, Laird MD, Dhandapani KM, Vender JR. Delayed reduction in hippocampal postsynaptic density protein-95 expression temporally correlates with cognitive dysfunction following controlled cortical impact in mice. J Neurosurg. 2010;113(6):1195–201.CrossRefPubMedCentralPubMed Wakade C, Sukumari-Ramesh S, Laird MD, Dhandapani KM, Vender JR. Delayed reduction in hippocampal postsynaptic density protein-95 expression temporally correlates with cognitive dysfunction following controlled cortical impact in mice. J Neurosurg. 2010;113(6):1195–201.CrossRefPubMedCentralPubMed
86.
go back to reference Han X, Tong J, Zhang J, Farahvar A, Wang E, Yang J, et al. Imipramine treatment improves cognitive outcome associated with enhanced hippocampal neurogenesis after traumatic brain injury in mice. J Neurotrauma. 2011;28(6):995–1007.CrossRefPubMedCentralPubMed Han X, Tong J, Zhang J, Farahvar A, Wang E, Yang J, et al. Imipramine treatment improves cognitive outcome associated with enhanced hippocampal neurogenesis after traumatic brain injury in mice. J Neurotrauma. 2011;28(6):995–1007.CrossRefPubMedCentralPubMed
87.
go back to reference Russo I, Caracciolo L, Tweedie D, Choi SH, Greig NH, Barlati S, et al. 3,6′-Dithiothalidomide, a new TNF-α synthesis inhibitor, attenuates the effect of Aβ1-42 intracerebroventricular injection on hippocampal neurogenesis and memory deficit. J Neurochem. 2012;122(6):1181–92.CrossRefPubMed Russo I, Caracciolo L, Tweedie D, Choi SH, Greig NH, Barlati S, et al. 3,6′-Dithiothalidomide, a new TNF-α synthesis inhibitor, attenuates the effect of Aβ1-42 intracerebroventricular injection on hippocampal neurogenesis and memory deficit. J Neurochem. 2012;122(6):1181–92.CrossRefPubMed
88.
go back to reference Belarbi K, Jopson T, Tweedie D, Arellano C, Luo W, Greig NH, et al. TNF-α protein synthesis inhibitor restores neuronal function and reverses cognitive deficits induced by chronic neuroinflammation. J Neuroinflammation. 2012;9:23.CrossRefPubMedCentralPubMed Belarbi K, Jopson T, Tweedie D, Arellano C, Luo W, Greig NH, et al. TNF-α protein synthesis inhibitor restores neuronal function and reverses cognitive deficits induced by chronic neuroinflammation. J Neuroinflammation. 2012;9:23.CrossRefPubMedCentralPubMed
89.
go back to reference Starke RM, Chalouhi N, Jabbour PM, Tjoumakaris SI, Gonzalez LF, Rosenwasser RH, et al. Critical role of TNF-α in cerebral aneurysm formation and progression to rupture. J Neuroinflammation. 2014;11:77.CrossRefPubMedCentralPubMed Starke RM, Chalouhi N, Jabbour PM, Tjoumakaris SI, Gonzalez LF, Rosenwasser RH, et al. Critical role of TNF-α in cerebral aneurysm formation and progression to rupture. J Neuroinflammation. 2014;11:77.CrossRefPubMedCentralPubMed
90.
go back to reference Ali MS, Starke RM, Jabbour PM, Tjoumakaris SI, Gonzalez LF, Rosenwasser RH, et al. TNF-α induces phenotypic modulation in cerebral vascular smooth muscle cells: implications for cerebral aneurysm pathology. J Cereb Blood Flow Metab. 2013;33(10):1564–73.CrossRefPubMedCentralPubMed Ali MS, Starke RM, Jabbour PM, Tjoumakaris SI, Gonzalez LF, Rosenwasser RH, et al. TNF-α induces phenotypic modulation in cerebral vascular smooth muscle cells: implications for cerebral aneurysm pathology. J Cereb Blood Flow Metab. 2013;33(10):1564–73.CrossRefPubMedCentralPubMed
91.
go back to reference Chio CC, Lin JW, Chang MW, Wang CC, Kuo JR, Yang CZ, et al. Therapeutic evaluation of etanercept in a model of traumatic brain injury. J Neurochem. 2010;115(4):921–9.CrossRefPubMed Chio CC, Lin JW, Chang MW, Wang CC, Kuo JR, Yang CZ, et al. Therapeutic evaluation of etanercept in a model of traumatic brain injury. J Neurochem. 2010;115(4):921–9.CrossRefPubMed
92.
go back to reference Lee CJ, Kim KW, Lee HM, Nahm FS, Lim YJ, Park JH, et al. The effect of thalidomide on spinal cord ischemia/reperfusion injury in a rabbit model. Spinal Cord. 2007;45(2):149–57.CrossRefPubMed Lee CJ, Kim KW, Lee HM, Nahm FS, Lim YJ, Park JH, et al. The effect of thalidomide on spinal cord ischemia/reperfusion injury in a rabbit model. Spinal Cord. 2007;45(2):149–57.CrossRefPubMed
93.
go back to reference Koopmans GC, Deumens R, Buss A, Geoghegan L, Myint AM, Honig WH, et al. Acute rolipram/thalidomide treatment improves tissue sparing and locomotion after experimental spinal cord injury. Exp Neurol. 2009;216(2):490–8.CrossRefPubMed Koopmans GC, Deumens R, Buss A, Geoghegan L, Myint AM, Honig WH, et al. Acute rolipram/thalidomide treatment improves tissue sparing and locomotion after experimental spinal cord injury. Exp Neurol. 2009;216(2):490–8.CrossRefPubMed
94.
go back to reference Genovese T, Mazzon E, Esposito E, Di Paola R, Caminiti R, Meli R, et al. Effect of thalidomide on signal transduction pathways and secondary damage in experimental spinal cord trauma. Shock. 2008;30(3):231–40.PubMed Genovese T, Mazzon E, Esposito E, Di Paola R, Caminiti R, Meli R, et al. Effect of thalidomide on signal transduction pathways and secondary damage in experimental spinal cord trauma. Shock. 2008;30(3):231–40.PubMed
96.
go back to reference Mahony C, Erskine L, Niven J, Greig NH, Figg WD, Vargesson N. Pomalidomide is nonteratogenic in chicken and zebrafish embryos and nonneurotoxic in vitro. Proc Natl Acad Sci U S A. 2013;110(31):12703–8.CrossRefPubMedCentralPubMed Mahony C, Erskine L, Niven J, Greig NH, Figg WD, Vargesson N. Pomalidomide is nonteratogenic in chicken and zebrafish embryos and nonneurotoxic in vitro. Proc Natl Acad Sci U S A. 2013;110(31):12703–8.CrossRefPubMedCentralPubMed
97.
go back to reference Vargesson N, Mahony C, Erskine L, Niven J, Greig NH, Figg WD. Reply to D’Amato et al. and Zeldis et al.: Screening of thalidomide derivatives in chicken and zebrafish embryos. Proc Natl Acad Sci U S A. 2013;110(50):E4820.CrossRefPubMedCentralPubMed Vargesson N, Mahony C, Erskine L, Niven J, Greig NH, Figg WD. Reply to D’Amato et al. and Zeldis et al.: Screening of thalidomide derivatives in chicken and zebrafish embryos. Proc Natl Acad Sci U S A. 2013;110(50):E4820.CrossRefPubMedCentralPubMed
98.
go back to reference Shohami E, Bass R, Wallach D, Yamin A, Gallily R. Inhibition of tumor necrosis factor alpha (TNFalpha) activity in rat brain is associated with cerebroprotection after closed head injury. J Cereb Blood Flow Metab. 1996;16(3):378–84.CrossRefPubMed Shohami E, Bass R, Wallach D, Yamin A, Gallily R. Inhibition of tumor necrosis factor alpha (TNFalpha) activity in rat brain is associated with cerebroprotection after closed head injury. J Cereb Blood Flow Metab. 1996;16(3):378–84.CrossRefPubMed
99.
go back to reference Sullivan PG, Bruce-Keller AJ, Rabchevsky AG, Christakos S, Clair DK, Mattson MP, et al. Exacerbation of damage and altered NF-kappaB activation in mice lacking tumor necrosis factor receptors after traumatic brain injury. J Neurosci. 1999;19:6248–56.PubMed Sullivan PG, Bruce-Keller AJ, Rabchevsky AG, Christakos S, Clair DK, Mattson MP, et al. Exacerbation of damage and altered NF-kappaB activation in mice lacking tumor necrosis factor receptors after traumatic brain injury. J Neurosci. 1999;19:6248–56.PubMed
100.
go back to reference Becker RE, Greig NH, Giacobini E, Schneider LS, Ferrucci L. A new roadmap for drug development for Alzheimer's disease. Nat Rev Drug Discov. 2014;13(2):156.CrossRefPubMed Becker RE, Greig NH, Giacobini E, Schneider LS, Ferrucci L. A new roadmap for drug development for Alzheimer's disease. Nat Rev Drug Discov. 2014;13(2):156.CrossRefPubMed
101.
go back to reference Ignatowski TA, Spengler RN, Dhandapani KM, Folkersma H, Butterworth RF, Tobinick E. Perispinal etanercept for post-stroke neurological and cognitive dysfunction: scientific rationale and current evidence. CNS Drugs. 2014;28:679–97.CrossRefPubMedCentralPubMed Ignatowski TA, Spengler RN, Dhandapani KM, Folkersma H, Butterworth RF, Tobinick E. Perispinal etanercept for post-stroke neurological and cognitive dysfunction: scientific rationale and current evidence. CNS Drugs. 2014;28:679–97.CrossRefPubMedCentralPubMed
102.
go back to reference Morris GP, Clark IA, Zinn R, Vissel B. Microglia: a new frontier for synaptic plasticity, learning and memory, and neurodegenerative disease research. Neurobiol Learn Mem. 2013;105:40–53.CrossRefPubMed Morris GP, Clark IA, Zinn R, Vissel B. Microglia: a new frontier for synaptic plasticity, learning and memory, and neurodegenerative disease research. Neurobiol Learn Mem. 2013;105:40–53.CrossRefPubMed
Metadata
Title
Transiently lowering tumor necrosis factor-α synthesis ameliorates neuronal cell loss and cognitive impairments induced by minimal traumatic brain injury in mice
Authors
Renana Baratz
David Tweedie
Jia-Yi Wang
Vardit Rubovitch
Weiming Luo
Barry J Hoffer
Nigel H Greig
Chaim G Pick
Publication date
01-12-2015
Publisher
BioMed Central
Published in
Journal of Neuroinflammation / Issue 1/2015
Electronic ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-015-0237-4

Other articles of this Issue 1/2015

Journal of Neuroinflammation 1/2015 Go to the issue