204
Views
5
CrossRef citations to date
0
Altmetric
Original Articles

The effects of glial cells inhibition on spatial reference, reversal and working memory deficits in a rat model of traumatic brain injury (TBI)

, , , &
Pages 226-236 | Received 20 Oct 2019, Accepted 29 Jul 2020, Published online: 19 Aug 2020

References

  • Syed AT, Lone NA, Wani MA, et al. Clinical management of patients with minor head injuries. Int J Health Sci (Qassim). 2007;1(1):131–140.
  • Marik PE, Varon J, Trask T. Management of head trauma. Chest. 2002;122(2):699–711.
  • Patt S, Brodhun M. Neuropathological sequelae of traumatic injury in the brain. An overview. Exp Toxicol Pathol. 1999;51(2):119–123.
  • Werner C, Engelhard K. Pathophysiology of traumatic brain injury. Br J Anaesth. 2007;99(1):4–9.
  • Karve IP, Taylor JM, Crack PJ. The contribution of astrocytes and microglia to traumatic brain injury. Br J Pharmacol. 2016;173(4):692–702.
  • McAllister TW. Neuropsychiatric sequelae of head injuries. Psychiatr Clin North Am. 1992;15(2):395–413.
  • Fields RD, Araque A, Johansen-Berg H, et al. Glial biology in learning and cognition. Neuroscientist. 2014;20(5):426–431.
  • Verkhratsky A, Butt A. Glial neurobiology: a textbook. John Wiley & Sons; 2007. p. 117.
  • Todd KJ, Serrano A, Lacaille J-C, et al. Glial cells in synaptic plasticity. J Physiol Paris. 2006;99(2-3):75–83.
  • Laird MD, Vender JR, Dhandapani KM. Opposing roles for reactive astrocytes following traumatic brain injury. Neurosignals. 2008;16(2-3):154–164.
  • Argaw AT, Asp L, Zhang J, Navrazhina K, et al. Astrocyte-derived VEGF-A drives blood-brain barrier disruption in CNS inflammatory disease. J Clin Invest. 2012;122(7):2454–2468.
  • Alvarez JI, Dodelet-Devillers A, Kebir H, et al. The hedgehog pathway promotes blood-brain barrier integrity and CNS immune quiescence. Science. 2011;334(6063):1727–1731.
  • Sofroniew MV, Vinters HV. Astrocytes: biology and pathology. Acta Neuropathol. 2010;119(1):7–35.
  • Bush TG, Puvanachandra N, Horner CH, et al. Leukocyte infiltration, neuronal degeneration, and neurite outgrowth after ablation of scar-forming, reactive astrocytes in adult transgenic mice. Neuron. 1999;23(2):297–308.
  • Fawcett JW, Asher RA. The glial scar and central nervous system repair. Brain Res Bull. 1999;49(6):377–391.
  • Giulian D. Reactive glia as rivals in regulating neuronal survival. Glia. 1993;7(1):102–110.
  • Horner PJ, Gage FH. Regenerating the damaged central nervous system. Nature. 2000;407(6807):963–970.
  • Silver J, Miller JH. Regeneration beyond the glial scar. Nat Rev Neurosci. 2004;5(2):146–156.
  • GrandPré T, Nakamura F, Vartanian T, et al. Identification of the Nogo inhibitor of axon regeneration as a Reticulon protein. Nature. 2000;403(6768):439–444.
  • Jones LL, Margolis RU, Tuszynski MH. The chondroitin sulfate proteoglycans neurocan, brevican, phosphacan, and versican are differentially regulated following spinal cord injury. Exp Neurol. 2003;182(2):399–411.
  • Moreau-Fauvarque C, Kumanogoh A, Camand E, et al. The transmembrane semaphorin Sema4D/CD100, an inhibitor of axonal growth, is expressed on oligodendrocytes and upregulated after CNS lesion. J Neurosci. 2003;23(27):9229–9239.
  • Shen LH, Li Y, Gao Q, et al. Down-regulation of neurocan expression in reactive astrocytes promotes axonal regeneration and facilitates the neurorestorative effects of bone marrow stromal cells in the ischemic rat brain . Glia. 2008;56(16):1747–1754.
  • Li Y, Chen J, Zhang CL, et al. Gliosis and brain remodeling after treatment of stroke in rats with marrow stromal cells. Glia. 2005;49(3):407–417.
  • Dixon CE, Clifton GL, Lighthall JW, et al. A controlled cortical impact model of traumatic brain injury in the rat. J Neurosci Methods. 1991;39(3):253–262.
  • Karimi SA, Hosseinmardi N, Janahmadi M, et al. The protective effect of hydrogen sulfide (H2S) on traumatic brain injury (TBI) induced memory deficits in rats. Brain Res Bull. 2017;134:177–182.
  • Sutton RL, Lescaudron L, Stein DG. Unilateral cortical contusion injury in the rat: vascular disruption and temporal development of cortical necrosis. J Neurotrauma. 1993;10(2):135–149.
  • Taylor AN, Rahman SU, Sanders NC, et al. Injury severity differentially affects short- and long-term neuroendocrine outcomes of traumatic brain injury. J Neurotrauma. 2008;25(4):311–323.
  • Wang L, Li C-C, Wang G-W, et al. The effects of centrally administered fluorocitrate via inhibiting glial cells on working memory in rats. Sci China, C, Life Sci. 2009;52(8):701–709.
  • Galgano M, Toshkezi G, Qiu X, et al. Traumatic brain injury: current treatment strategies and future endeavors. Cell Transplant. 2017;26(7):1118–1130.
  • Simon-O'Brien E, Gauthier D, Riban V, et al. Etifoxine improves sensorimotor deficits and reduces glial activation, neuronal degeneration, and neuroinflammation in a rat model of traumatic brain injury. J Neuroinflammation. 2016;13(1):203.
  • Dixon CE, Lyeth BG, Povlishock JT, et al. A fluid percussion model of experimental brain injury in the rat. J Neurosurg. 1987;67(1):110–119.
  • Hamm RJ, Dixon CE, Gbadebo DM, et al. Cognitive deficits following traumatic brain injury produced by controlled cortical impact. J Neurotrauma. 1992;9(1):11–20.
  • Karimi SA, Salehi I, Shykhi T, et al. Effects of exposure to extremely low-frequency electromagnetic fields on spatial and passive avoidance learning and memory, anxiety-like behavior and oxidative stress in male rats. Behav Brain Res. 2019;359:630–638.
  • Gholamipour-Badie H, Naderi N, Khodagholi F, et al. L-type calcium channel blockade alleviates molecular and reversal spatial learning and memory alterations induced by entorhinal amyloid pathology in rats. Behav Brain Res. 2013;237:190–199.
  • Nategh M, Nikseresht S, Khodagholi F, et al. Physiology& behavior nucleus incertus inactivation impairs spatial learning and memory in rats. Physiol Behav. 2015;139:112–120.
  • Hassel B, Paulsen RE, Johnsen A, et al. Selective inhibition of glial cell metabolism in vivo by fluorocitrate. Brain Res. 1992;576(1):120–124.
  • Hassel B, Sonnewald U, Unsgård G, et al. NMR spectroscopy of cultured astrocytes: effects of glutamine and the gliotoxin fluorocitrate. J Neurochem. 1994;62(6):2187–2194.,
  • Kaur P, Sharma S. Recent advances in pathophysiology of traumatic brain injury. Curr Neuropharmacol. 2018;16(8):1224–1238.
  • Xiong Y, Mahmood A, Chopp M. Emerging treatments for traumatic brain injury. Expert Opin Emerg Drugs. 2009;14(1):67–84.
  • Buffo A, Rolando C, Ceruti S. Astrocytes in the damaged brain: molecular and cellular insights into their reactive response and healing potential. Biochem Pharmacol. 2010;79(2):77–89.
  • Burda JE, Bernstein AM, Sofroniew MV. Astrocyte roles in traumatic brain injury. Exp Neurol. 2016;275:305–315.
  • Kimelberg HK. Astrocytic swelling in cerebral ischemia as a possible cause of injury and target for therapy. Glia. 2005;50(4):389–397.
  • Loane DJ, Kumar A. Microglia in the TBI brain: The good, the bad, and the dysregulated. Exp Neurol. 2016;275 Pt 3:316–327.
  • Kettenmann H, Hanisch U-K, Noda M, et al. Physiology of microglia. Physiol Rev. 2011;91(2):461–553.
  • Loane DJ, Kumar A, Stoica BA, et al. Progressive neurodegeneration after experimental brain trauma: association with chronic microglial activation. J Neuropathol Exp Neurol. 2014;73(1):14–29.
  • Myer DJ, Gurkoff GG, Lee SM, et al. Essential protective roles of reactive astrocytes in traumatic brain injury. Brain. 2006;129(Pt 10):2761–2772.
  • Christopherson KS, Ullian EM, Stokes CCA, et al. Thrombospondins are astrocyte-secreted proteins that promote CNS synaptogenesis. Cell. 2005;120(3):421–433.,.
  • Yi JH, Katagiri Y, Susarla B, et al. Alterations in sulfated chondroitin glycosaminoglycans following controlled cortical impact injury in mice. J Comp Neurol. 2012;520(15):3295–3313.
  • Shohami E, Ginis I, Hallenbeck JM. Dual role of tumor necrosis factor alpha in brain injury. Cytokine Growth Factor Rev. 1999;10.
  • Cederberg D, Siesjö P. What has inflammation to do with traumatic brain injury? Childs Nerv Syst. 2010;26(2):221–226.
  • Largo C, Cuevas P, Somjen GG, et al. The effect of depressing glial function in rat brain in situ on ion homeostasis, synaptic transmission, and neuron survival. J Neurosci. 1996;16(3):1219–1229.,
  • Hirose S, Umetani Y, Amitani M, et al. Role of NMDA receptors in the increase of glucose metabolism in the rat brain induced by fluorocitrate. Neurosci Lett. 2007;415(3):259–263.
  • Mothet JP, Rouaud E, Sinet PM, et al. A critical role for the glial-derived neuromodulator d-serine in the age-related deficits of cellular mechanisms of learning and memory. Aging Cell. 2006;5(3):267–274.
  • Adamsky A, Goshen I. Astrocytes in memory function: pioneering findings and future directions. Neuroscience. 2018;370:14–26.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.