Skip to main content

Advertisement

Log in

Disruption of Ion Homeostasis in the Neurogliovascular Unit Underlies the Pathogenesis of Ischemic Cerebral Edema

  • Original Article
  • Published:
Translational Stroke Research Aims and scope Submit manuscript

Abstract

Cerebral edema is a major cause of morbidity and mortality following ischemic stroke, but its underlying molecular pathophysiology is incompletely understood. Recent data have revealed the importance of ion flux via channels and transporters expressed in the neurogliovascular unit in the development of ischemia-triggered cytotoxic edema, vasogenic edema, and hemorrhagic conversion. Disruption of homeostatic mechanisms governing cell volume regulation and epithelial/endothelial ion transport due to ischemia-associated energy failure results in the thermodynamically driven re-equilibration of solutes and water across the CSF–blood and blood–brain barriers that ultimately increases the brain’s extravascular volume. Additionally, hypoxia, inflammation, and other stress-triggered increases in the functional expression of ion channels and transporters normally expressed at low levels in the neurogliovascular unit cause disruptions in ion homeostasis that contribute to ischemic cerebral edema. Here, we review the pathophysiological significance of several molecular mediators of ion transport expressed in the neurogliovascular unit, including targets of existing FDA-approved drugs, which might be potential nodes for therapeutic intervention.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3

Similar content being viewed by others

References

  1. Luissint A-C et al. Tight junctions at the blood brain barrier: physiological architecture and disease-associated dysregulation. Fluids Barriers CNS. 2012;9(1):23.

    PubMed Central  PubMed  Google Scholar 

  2. Liu W-Y et al. Tight junction in blood–brain barrier: an overview of structure, regulation, and regulator substances. CNS Neurosci Ther. 2012;18(8):609–15.

    CAS  PubMed  Google Scholar 

  3. Abbott NJ et al. Structure and function of the blood–brain barrier. Neurobiol Dis. 2010;37(1):13–25.

    CAS  PubMed  Google Scholar 

  4. Nag, S. Morphology and properties of astrocytes. In: The blood–brain and other neural barriers, S. Nag, editor; 2011. Totowa: Humana. p. 69–100.

  5. Wolburg H et al. Brain endothelial cells and the glio-vascular complex. Cell Tissue Res. 2009;335(1):75–96.

    PubMed  Google Scholar 

  6. Abbott NJ, Ronnback L, Hansson E. Astrocyte-endothelial interactions at the blood–brain barrier. Nat Rev Neurosci. 2006;7(1):41–53.

    CAS  PubMed  Google Scholar 

  7. Lassmann H et al. Microglial cells are a component of the perivascular glia limitans. J Neurosci Res. 1991;28(2):236–43.

    CAS  PubMed  Google Scholar 

  8. Sá-Pereira I, Brites D, Brito M. Neurovascular unit: a focus on pericytes. Mol Neurobiol. 2012;45(2):327–47.

    PubMed  Google Scholar 

  9. Simard JM et al. Brain oedema in focal ischaemia: molecular pathophysiology and theoretical implications. Lancet Neurol. 2007;6(3):258–68.

    CAS  PubMed Central  PubMed  Google Scholar 

  10. Betz AL. Alterations in cerebral endothelial cell function in ischemia. Adv Neurol. 1996;71:301–11. discussion 311–3.

    CAS  PubMed  Google Scholar 

  11. Menzies, S.A., J.T. Hoff, and A.L. Betz. Extravasation of albumin in ischaemic brain oedema. In: Brain edema VIII. H.-J. Reulen, et al., editors; 1990. Vienna: Springer. p. 220–2.

  12. Schielke GP, Moises HC, Betz AL. Blood to brain sodium transport and interstitial fluid potassium concentration during early focal ischemia in the rat. J Cereb Blood Flow Metab. 1991;11(3):466–71.

    CAS  PubMed  Google Scholar 

  13. Rajasekaran SA, Beyenbach KW, Rajasekaran AK. Interactions of tight junctions with membrane channels and transporters. Biochim Biophys Acta. 2008;1778(3):757–69.

    CAS  PubMed  Google Scholar 

  14. Menzies SA, Betz AL, Hoff JT. Contributions of ions and albumin to the formation and resolution of ischemic brain edema. J Neurosurg. 1993;78(2):257–66.

    CAS  PubMed  Google Scholar 

  15. Wu Q et al. Functional demonstration of Na+−K+−2Cl cotransporter activity in isolated, polarized choroid plexus cells. Am J Physiol - Cell Physiol. 1998;275(6):C1565–72.

    CAS  Google Scholar 

  16. Kahle KT et al. Roles of the cation–chloride cotransporters in neurological disease. Nat Clin Pract Neurol. 2008;4(9):490–503.

    CAS  PubMed  Google Scholar 

  17. Kang T-C et al. Changes in Na+−K+−Cl cotransporter immunoreactivity in the gerbil hippocampus following transient ischemia. Neurosci Res. 2002;44(3):249–54.

    CAS  PubMed  Google Scholar 

  18. Yan Y, Dempsey RJ, Sun D. Na+−K+−Cl cotransporter in rat focal cerebral ischemia. J Cereb Blood Flow Metab. 2001;21(6):711–21.

    CAS  PubMed  Google Scholar 

  19. Yan Y et al. Inhibition of Na+−K+−Cl cotransporter during focal cerebral ischemia decreases edema and neuronal damage. Brain Res. 2003;961(1):22–31.

    CAS  PubMed  Google Scholar 

  20. Dai Y, Tang J, Zhang JH. Role of Cl in cerebral vascular tone and expression of Na+−K+−2Cl co-transporter after neonatal hypoxia–ischemia. Can J Physiol Pharmacol. 2005;83(8–9):767–73.

    CAS  PubMed  Google Scholar 

  21. Lee H-A et al. Possible involvement of DNA methylation in NKCC1 gene expression during postnatal development and in response to ischemia. J Neurochem. 2010;114(2):520–9.

    CAS  PubMed  Google Scholar 

  22. Lenart B et al. Na–K–Cl cotransporter-mediated intracellular Na+ accumulation affects Ca2+ signaling in astrocytes in an in vitro ischemic model. J Neurosci. 2004;24(43):9585–97.

    CAS  PubMed  Google Scholar 

  23. Foroutan S et al. Moderate-to-severe ischemic conditions increase activity and phosphorylation of the cerebral microvascular endothelial cell Na+−K+−Cl cotransporter. Am J Physiol—Cell Physiol. 2005;289(6):C1492–501.

    CAS  PubMed  Google Scholar 

  24. Geng Y, Hoke A, Delpire E. The Ste20 kinases Ste20-related proline-alanine-rich kinase and oxidative-stress response 1 regulate NKCC1 function in sensory neurons. J Biol Chem. 2009;284(21):14020–8.

    CAS  PubMed  Google Scholar 

  25. Kahle, K.T., J. Rinehart, and R.P. Lifton. Phosphoregulation of the Na–K–2Cl and K–Cl cotransporters by the WNK kinases. Biochimica et Biophysica Acta (BBA)—Molecular Basis of Disease, 2010; 1802(12):1150–8.

    Google Scholar 

  26. Sun D, Murali SG. Stimulation of Na+−K+−2Cl cotransporter in neuronal cells by excitatory neurotransmitter glutamate. Am J Physiol—Cell Physiol. 1998;275(3):C772–9.

    CAS  Google Scholar 

  27. Beck J et al. Na-K–Cl cotransporter contributes to glutamate-mediated excitotoxicity. J Neurosci. 2003;23(12):5061–8.

    CAS  PubMed  Google Scholar 

  28. Su G, Kintner DB, Sun D. Contribution of Na+−K+−Cl cotransporter to high-[K+]o-induced swelling and EAA release in astrocytes. Am J Physiol—Cell Physiol. 2002;282(5):C1136–46.

    CAS  PubMed  Google Scholar 

  29. Su G et al. Astrocytes from Na+−K+−Cl cotransporter-null mice exhibit absence of swelling and decrease in EAA release. Am J Physiol—Cell Physiol. 2002;282(5):C1147–60.

    CAS  PubMed  Google Scholar 

  30. Schomberg SL et al. Stimulation of Na–K–2Cl cotransporter in neurons by activation of non-NMDA ionotropic receptor and group-I mGluRs. J Neurophysiol. 2001;85(6):2563–75.

    CAS  PubMed  Google Scholar 

  31. Sun D, Lytle C, O’Donnell ME. IL-6 secreted by astroglial cells regulates Na–K–Cl cotransport in brain microvessel endothelial cells. Am J Physiol—Cell Physiol. 1997;272(6):C1829–35.

    CAS  Google Scholar 

  32. Chen H et al. Na+-dependent chloride transporter (NKCC1)-null mice exhibit less gray and white matter damage after focal cerebral ischemia. J Cereb Blood Flow Metab. 2005;25(1):54–66.

    PubMed  Google Scholar 

  33. Chen H, Sun D. The role of Na–K–Cl co-transporter in cerebral ischemia. Neurol Res. 2005;27(3):280–6.

    CAS  PubMed  Google Scholar 

  34. O’Donnell ME et al. Bumetanide inhibition of the blood–brain barrier Na–K–Cl cotransporter reduces edema formation in the rat middle cerebral artery occlusion model of stroke. J Cereb Blood Flow Metab. 2004;24(9):1046–56.

    PubMed  Google Scholar 

  35. McClain RM, Dammers KD. Toxicologic evaluation of bumetanide, a potent diuretic agent. J Clin Pharmacol. 1981;21(11):543–54.

    CAS  PubMed  Google Scholar 

  36. Flamenbaum W, Friedman R. Pharmacology, therapeutic efficacy, and adverse effects of bumetanide, a new “loop” diuretic. Pharmacotherapy: J Human Pharmacol Drug Ther. 1982;2(4):213–22.

    CAS  Google Scholar 

  37. Khanna, A., B.P. Walcott, and K.T. Kahle. Limitations of current GABA agonists in neonatal seizures: towards GABA modulation via the targeting of neuronal Cl transport. Front Neurol, 2013; 4:78.

    Google Scholar 

  38. Li Y et al. Sensitive isotope dilution liquid chromatography/tandem mass spectrometry method for quantitative analysis of bumetanide in serum and brain tissue. J Chromatogr B Analyt Technol Biomed Life Sci. 2011;879(13–14):998–1002.

    CAS  PubMed Central  PubMed  Google Scholar 

  39. Payne JA. Functional characterization of the neuronal-specific K–Cl cotransporter: implications for [K+]o regulation. Am J Physiol. 1997;273(5 Pt 1):C1516–25.

    CAS  PubMed  Google Scholar 

  40. Kahle KT et al. Molecular mechanisms of ischemic cerebral edema: role of electroneutral ion transport. Physiology. 2009;24(4):257–65.

    CAS  PubMed  Google Scholar 

  41. Launay P et al. TRPM4 is a Ca2+-activated nonselective cation channel mediating cell membrane depolarization. Cell. 2002;109(3):397–407.

    CAS  PubMed  Google Scholar 

  42. Xu X-ZS et al. Regulation of melastatin, a TRP-related protein, through interaction with a cytoplasmic isoform. Proc Natl Acad Sci. 2001;98(19):10692–7.

    CAS  PubMed  Google Scholar 

  43. Woo SK et al. The sulfonylurea receptor 1 (Sur1)-transient receptor potential melastatin 4 (Trpm4) channel. J Biol Chem. 2013;288(5):3655–67.

    CAS  PubMed  Google Scholar 

  44. Simard JM et al. Newly expressed SUR1-regulated NC(Ca-ATP) channel mediates cerebral edema after ischemic stroke. Nat Med. 2006;12(4):433–40.

    CAS  PubMed Central  PubMed  Google Scholar 

  45. Chen M, Dong Y, Simard JM. Functional coupling between sulfonylurea receptor type 1 and a nonselective cation channel in reactive astrocytes from adult rat brain. J Neurosci. 2003;23(24):8568–77.

    CAS  PubMed  Google Scholar 

  46. Gerzanich V et al. De novo expression of Trpm4 initiates secondary hemorrhage in spinal cord injury. Nat Med. 2009;15(2):185–91.

    CAS  PubMed Central  PubMed  Google Scholar 

  47. Simard JM, Tarasov KV, Gerzanich V. Non-selective cation channels, transient receptor potential channels and ischemic stroke. Biochim Biophys Acta. 2007;1772(8):947–57.

    CAS  PubMed Central  PubMed  Google Scholar 

  48. Simard JM et al. Drugs acting on SUR1 to treat CNS ischemia and trauma. Curr Opin Pharmacol. 2008;8(1):42–9.

    CAS  PubMed Central  PubMed  Google Scholar 

  49. Chen M, Simard JM. Cell swelling and a nonselective cation channel regulated by internal Ca2+ and ATP in native reactive astrocytes from adult rat brain. J Neurosci. 2001;21(17):6512–21.

    CAS  PubMed  Google Scholar 

  50. Simard JM et al. Endothelial sulfonylurea receptor 1-regulated NC Ca-ATP channels mediate progressive hemorrhagic necrosis following spinal cord injury. J Clin Invest. 2007;117(8):2105–13.

    CAS  PubMed Central  PubMed  Google Scholar 

  51. Simard JM et al. Protective effect of delayed treatment with low-dose glibenclamide in three models of ischemic stroke. Stroke. 2009;40(2):604–9.

    CAS  PubMed Central  PubMed  Google Scholar 

  52. Wu X, Reddy DS. Integrins as receptor targets for neurological disorders. Pharmacol Ther. 2012;134(1):68–81.

    CAS  PubMed Central  PubMed  Google Scholar 

  53. Simard JM et al. Glibenclamide reduces inflammation, vasogenic edema, and caspase-3 activation after subarachnoid hemorrhage. J Cereb Blood Flow Metab. 2009;29(2):317–30.

    CAS  PubMed Central  PubMed  Google Scholar 

  54. Simard JM et al. Does inhibiting Sur1 complement rt-PA in cerebral ischemia? Ann N Y Acad Sci. 2012;1268:95–107.

    CAS  PubMed Central  PubMed  Google Scholar 

  55. Ortega FJ et al. ATP-dependent potassium channel blockade strengthens microglial neuroprotection after hypoxia–ischemia in rats. Exp Neurol. 2012;235(1):282–96.

    CAS  PubMed  Google Scholar 

  56. Simard JM et al. Glibenclamide—10-h treatment window in a clinically relevant model of stroke. Transl Stroke Res. 2012;3(2):286–95.

    CAS  PubMed Central  PubMed  Google Scholar 

  57. Wali B et al. Glibenclamide administration attenuates infarct volume, hemispheric swelling, and functional impairments following permanent focal cerebral ischemia in rats. Stroke Res Treat. 2012;2012:6.

    Google Scholar 

  58. Lima FO et al. The pattern of leptomeningeal collaterals on CT angiography is a strong predictor of long-term functional outcome in stroke patients with large vessel intracranial occlusion. Stroke. 2010;41(10):2316–22.

    PubMed  Google Scholar 

  59. Simard JM et al. Glibenclamide is superior to decompressive craniectomy in a rat model of malignant stroke. Stroke. 2010;41(3):531–7.

    CAS  PubMed Central  PubMed  Google Scholar 

  60. Moretti ME, Rezvani M, Koren G. Safety of glyburide for gestational diabetes: a meta-analysis of pregnancy outcomes. Ann Pharmacother. 2008;42(4):483–90.

    CAS  PubMed  Google Scholar 

  61. Rendell M. The role of sulphonylureas in the management of type 2 diabetes mellitus. Drugs. 2004;64(12):1339–58.

    CAS  PubMed  Google Scholar 

  62. Kunte H et al. Sulfonylureas improve outcome in patients with type 2 diabetes and acute ischemic stroke. Stroke. 2007;38(9):2526–30.

    CAS  PubMed Central  PubMed  Google Scholar 

  63. Boron WF. Regulation of intracellular pH. Adv Physiol Educ. 2004;28(4):160–79.

    PubMed  Google Scholar 

  64. Mahnensmith RL, Aronson PS. The plasma membrane sodium-hydrogen exchanger and its role in physiological and pathophysiological processes. Circ Res. 1985;56(6):773–88.

    CAS  PubMed  Google Scholar 

  65. Pedersen SF et al. Physiology and pathophysiology of Na+/H+ exchange and Na+−K+−2Cl cotransport in the heart, brain, and blood. Am J Physiol Regul Integr Comp Physiol. 2006;291(1):R1–R25.

    CAS  PubMed  Google Scholar 

  66. Orlowski J, Grinstein S. Na+/H+ exchangers of mammalian cells. J Biol Chem. 1997;272(36):22373–6.

    CAS  PubMed  Google Scholar 

  67. Nakamura N et al. Four Na+/H+ exchanger isoforms are distributed to Golgi and post-Golgi compartments and are involved in organelle pH regulation. J Biol Chem. 2005;280(2):1561–72.

    CAS  PubMed  Google Scholar 

  68. Orlowski J, Kandasamy RA, Shull GE. Molecular cloning of putative members of the Na/H exchanger gene family. cDNA cloning, deduced amino acid sequence, and mRNA tissue expression of the rat Na/H exchanger NHE-1 and two structurally related proteins. J Biol Chem. 1992;267(13):9331–9.

    CAS  PubMed  Google Scholar 

  69. Chesler M. Regulation and modulation of pH in the brain. Physiol Rev. 2003;83(4):1183–221.

    CAS  PubMed  Google Scholar 

  70. Szabó EZ et al. Kinetic and pharmacological properties of human brain Na+/H+ exchanger isoform 5 stably expressed in Chinese hamster ovary cells. J Biol Chem. 2000;275(9):6302–7.

    PubMed  Google Scholar 

  71. Noel J, Roux D, Pouyssegur J. Differential localization of Na+/H+ exchanger isoforms (NHE1 and NHE3) in polarized epithelial cell lines. J Cell Sci. 1996;109(5):929–39.

    CAS  PubMed  Google Scholar 

  72. Wang XF et al. Expression, immunolocalization, and functional activity of Na+/H+ exchanger isoforms in mouse endometrial epithelium. Biol Reprod. 2003;68(1):302–8.

    CAS  PubMed  Google Scholar 

  73. Sipos I et al. Impaired regulation of ph homeostasis by oxidative stress in rat brain capillary endothelial cells. Cell Mol Neurobiol. 2005;25(1):141–51.

    CAS  PubMed  Google Scholar 

  74. Hsu P et al. pHi in piglet cerebral microvascular endothelial cells: recovery from an acid load. Exp Biol Med. 1996;212(3):256–62.

    CAS  Google Scholar 

  75. Lam TI, Wise PM, O’Donnell ME. Cerebral microvascular endothelial cell Na/H exchange: evidence for the presence of NHE1 and NHE2 isoforms and regulation by arginine vasopressin. Am J Physiol—Cell Physiol. 2009;297(2):C278–89.

    CAS  PubMed  Google Scholar 

  76. Hom S et al. Comparative changes in the blood–brain barrier and cerebral infarction of SHR and WKY rats. Am J Physiol Regul Integr Comp Physiol. 2007;292(5):R1881–92.

    CAS  PubMed  Google Scholar 

  77. Kawai, N., R.M. McCarron, and M. Spatz. The effect of endothelins on ion transport systems in cultured rat brain capillary endothelial cells. In: Brain edema X, H. James, et al., editors; 1997. Vienna: Springer. p. 138–40.

  78. Vigne P, Ladoux A, Frelin C. Endothelins activate Na+/H+ exchange in brain capillary endothelial cells via a high affinity endothelin-3 receptor that is not coupled to phospholipase C. J Biol Chem. 1991;266(9):5925–8.

    CAS  PubMed  Google Scholar 

  79. Luo J et al. Decreased neuronal death in Na+/H+ exchanger isoform 1-null mice after in vitro and in vivo ischemia. J Neurosci. 2005;25(49):11256–68.

    CAS  PubMed  Google Scholar 

  80. Betz AL. Sodium transport from blood to brain: inhibition by furosemide and amiloride. J Neurochem. 1983;41(4):1158–64.

    CAS  PubMed  Google Scholar 

  81. Ennis SR, Ren X-D, Betz AL. Mechanisms of sodium transport at the blood–brain barrier studied with in situ perfusion of rat brain. J Neurochem. 1996;66(2):756–63.

    CAS  PubMed  Google Scholar 

  82. Horikawa N et al. Na+/H+ exchange inhibitor SM-20220 improves endothelial dysfunction induced by ischemia–reperfusion. Jpn J Pharmacol. 2001;85(3):271–7.

    CAS  PubMed  Google Scholar 

  83. Horikawa N et al. Relationship between the neuroprotective effect of Na+/H+ exchanger inhibitor SM-20220 and the timing of its administration in a transient middle cerebral artery occlusion model of rats. Biol Pharm Bull. 2001;24(7):767–71.

    CAS  PubMed  Google Scholar 

  84. Kitayama J et al. Inhibition of Na+/H+ exchanger reduces infarct volume of focal cerebral ischemia in rats. Brain Res. 2001;922(2):223–8.

    CAS  PubMed  Google Scholar 

  85. Ferrazzano P et al. Inhibiting the Na+/H+ exchanger reduces reperfusion injury: a small animal MRI study. Front Biosci (Elite Ed). 2011;1(3):81–8.

    Google Scholar 

  86. O’Donnell ME et al. Intravenous HOE-642 reduces brain edema and Na uptake in the rat permanent middle cerebral artery occlusion model of stroke: evidence for participation of the blood–brain barrier Na/H exchanger. J Cereb Blood Flow Metab. 2013;33(2):225–34.

    PubMed  Google Scholar 

  87. Castella M, Buckberg GD, Tan Z. Neurologic preservation by Na+−H+ exchange inhibition prior to 90 minutes of hypothermic circulatory arrest. Ann Thorac Surg. 2005;79(2):646–54.

    PubMed  Google Scholar 

  88. Masereel B, Pochet L, Laeckmann D. An overview of inhibitors of Na+/H+ exchanger. Eur J Med Chem. 2003;38(6):547–54.

    CAS  PubMed  Google Scholar 

  89. Szatkowski M, Attwell D. Triggering and execution of neuronal death in brain ischaemia: two phases of glutamate release by different mechanisms. Trends Neurosci. 1994;17(9):359–65.

    CAS  PubMed  Google Scholar 

  90. Rothman SM, Olney JW. Glutamate and the pathophysiology of hypoxic–ischemic brain damage. Ann Neurol. 1986;19(2):105–11.

    CAS  PubMed  Google Scholar 

  91. Rothman SM, Olney JW. Excitotoxicity and the NMDA receptor—still lethal after eight years. Trends Neurosci. 1995;18(2):57–8.

    CAS  PubMed  Google Scholar 

  92. Choi DW. Excitotoxic cell death. J Neurobiol. 1992;23(9):1261–76.

    CAS  PubMed  Google Scholar 

  93. Sattler R, Tymianski M. Molecular mechanisms of glutamate receptor-mediated excitotoxic neuronal cell death. Mol Neurobiol. 2001;24(1–3):107–29.

    CAS  PubMed  Google Scholar 

  94. Kristián T, Siesjö BK. Calcium in ischemic cell death. Stroke. 1998;29(3):705–18.

    PubMed  Google Scholar 

  95. Choi D, Maulucci-Gedde M, Kriegstein A. Glutamate neurotoxicity in cortical cell culture. J Neurosci. 1987;7(2):357–68.

    CAS  PubMed  Google Scholar 

  96. Rothman S. The neurotoxicity of excitatory amino acids is produced by passive chloride influx. J Neurosci. 1985;5(6):1483–9.

    CAS  PubMed  Google Scholar 

  97. Hansson E et al. Astroglia and glutamate in physiology and pathology: aspects on glutamate transport, glutamate-induced cell swelling and gap-junction communication. Neurochem Int. 2000;37(2–3):317–29.

    CAS  PubMed  Google Scholar 

  98. Sharp C et al. Human neuroepithelial cells express NMDA receptors. BMC Neurosci. 2003;4(1):28.

    PubMed Central  PubMed  Google Scholar 

  99. Sharp CD et al. Glutamate causes a loss in human cerebral endothelial barrier integrity through activation of NMDA receptor. Am J Physiol Heart Circ Physiol. 2003;285(6):H2592–8.

    CAS  PubMed  Google Scholar 

  100. Westergren I, Johansson BB. Blockade of AMPA receptors reduces brain edema following opening of the blood–brain barrier. J Cereb Blood Flow Metab. 1993;13(4):603–8.

    CAS  PubMed  Google Scholar 

  101. Minematsu K et al. Effects of a novel NMDA antagonist on experimental stroke rapidly and quantitatively assessed by diffusion-weighted MRI. Neurology. 1993;43(2):397.

    CAS  PubMed  Google Scholar 

  102. Westergren I, Johansson BB. NBQX, an AMPA antagonist, reduces glutamate-mediated brain edema. Brain Res. 1992;573(2):324–6.

    CAS  PubMed  Google Scholar 

  103. Ikonomidou C, Turski L. Why did NMDA receptor antagonists fail clinical trials for stroke and traumatic brain injury? Lancet Neurol. 2002;1(6):383–6.

    CAS  PubMed  Google Scholar 

  104. Besancon E et al. Beyond NMDA and AMPA glutamate receptors: emerging mechanisms for ionic imbalance and cell death in stroke. Trends Pharmacol Sci. 2008;29(5):268–75.

    CAS  PubMed  Google Scholar 

  105. Chen H et al. Open-channel block of N-methyl-d-aspartate (NMDA) responses by memantine: therapeutic advantage against NMDA receptor-mediated neurotoxicity. J Neurosci. 1992;12(11):4427–36.

    CAS  PubMed  Google Scholar 

  106. Tozzi A et al. Memantine reduces neuronal dysfunctions triggered by in vitro ischemia and 3-nitropropionic acid. Exp Neurol. 2007;207(2):218–26.

    CAS  PubMed  Google Scholar 

  107. Görgülü A et al. Reduction of edema and infarction by memantine and MK-801 after focal cerebral ischaemia and reperfusion in rat. Acta Neurochir. 2000;142(11):1287–92.

    PubMed  Google Scholar 

  108. Mdzinarishvili, A., et al.. NGP1-01, a lipophilic polycyclic cage amine, is neuroprotective in focal ischemia. Neurosci Lett. 2005;383(1–2):49–53.

    Google Scholar 

  109. Li J et al. The pre-ischaemic neuroprotective effect of a novel polyamine antagonist, N1-dansyl-spermine in a permanent focal cerebral ischaemia model in mice. Brain Res. 2004;1029(1):84–92.

    CAS  PubMed  Google Scholar 

  110. Diarra A et al. Anoxia-evoked intracellular pH and Ca2+ concentration changes in cultured postnatal rat hippocampal neurons. Neuroscience. 1999;93(3):1003–16.

    CAS  PubMed  Google Scholar 

  111. Allen NJ, Attwell D. Modulation of ASIC channels in rat cerebellar purkinje neurons by ischaemia-related signals. J Physiol. 2002;543(2):521–9.

    CAS  PubMed  Google Scholar 

  112. Obrenovitch TP et al. A rapid redistribution of hydrogen ions is associated with depolarization and repolarization subsequent to cerebral ischemia reperfusion. J Neurophysiol. 1990;64(4):1125–33.

    CAS  PubMed  Google Scholar 

  113. Waldmann R et al. H+-gated cation channels. Ann N Y Acad Sci. 1999;868(1):67–76.

    CAS  PubMed  Google Scholar 

  114. Waldmann R, Lazdunski M. H+-gated cation channels: neuronal acid sensors in the NaC/DEG family of ion channels. Curr Opin Neurobiol. 1998;8(3):418–24.

    CAS  PubMed  Google Scholar 

  115. Wemmie JA et al. Acid-sensing ion channel 1 is localized in brain regions with high synaptic density and contributes to fear conditioning. J Neurosci. 2003;23(13):5496–502.

    CAS  PubMed  Google Scholar 

  116. Alvarez de la Rosa D et al. Distribution, subcellular localization and ontogeny of ASIC1 in the mammalian central nervous system. J Physiol. 2003;546(Pt 1):77–87.

    CAS  PubMed  Google Scholar 

  117. Yermolaieva O et al. Extracellular acidosis increases neuronal cell calcium by activating acid-sensing ion channel 1a. Proc Natl Acad Sci U S A. 2004;101(17):6752–7.

    CAS  PubMed Central  PubMed  Google Scholar 

  118. Nedergaard M et al. Dynamics of interstitial and intracellular pH in evolving brain infarct. Am J Physiol Regul Integr Comp Physiol. 1991;260(3):R581–8.

    CAS  Google Scholar 

  119. Rehncrona S. Brain acidosis. Ann Emerg Med. 1985;14(8):770–6.

    CAS  PubMed  Google Scholar 

  120. Smith, M.-L. and B. Siesjö. Acidosis-related brain damage: immediate and delayed events. In: Mechanisms of cerebral hypoxia and stroke, G. Somjen, editor; 1988. New York: Springer. p. 57–71.

  121. Xiong Z-G et al. Neuroprotection in ischemia: blocking calcium-permeable acid-sensing ion channels. Cell. 2004;118(6):687–98.

    CAS  PubMed  Google Scholar 

  122. Johnson MB et al. Global ischemia induces expression of acid-sensing ion channel 2a in rat brain. J Cereb Blood Flow Metab. 2001;21(6):734–40.

    CAS  PubMed  Google Scholar 

  123. Chen X, Kalbacher H, Gründer S. The tarantula toxin psalmotoxin 1 inhibits acid-sensing ion channel (ASIC) 1a by increasing its apparent H+ affinity. J Gen Physiol. 2005;126(1):71–9.

    CAS  PubMed Central  PubMed  Google Scholar 

  124. Voilley N et al. Nonsteroid anti-inflammatory drugs inhibit both the activity and the inflammation-induced expression of acid-sensing ion channels in nociceptors. J Neurosci. 2001;21(20):8026–33.

    CAS  PubMed  Google Scholar 

  125. Chu X-P et al. Subunit-dependent high-affinity zinc inhibition of acid-sensing ion channels. J Neurosci. 2004;24(40):8678–89.

    CAS  PubMed Central  PubMed  Google Scholar 

  126. Gees, M., et al.. TRP channels. Comprehensive Physiology; 2012.

  127. Woodard, G.E., S.O. Sage, and J.A. Rosado. Transient receptor potential channels and intracellular signaling. In: International review of cytology, W.J. Kwang, editor; 2007. New York: Academic. p. 35–67.

  128. Simard JM, Tarasov KV, Gerzanich V. Non-selective cation channels, transient receptor potential channels and ischemic stroke. Biochim Biophys Acta (BBA)—Mol Basis Dis. 2007;1772(8):947–57.

    CAS  Google Scholar 

  129. Fonfria E et al. TRPM2 is elevated in the tMCAO stroke model, transcriptionally regulated, and functionally expressed in C13 microglia. J Receptors Signal Transduct. 2006;26(3):179–98.

    CAS  Google Scholar 

  130. Gao Y et al. [Expression of transient receptor potential channel 4 in striatum and hippocampus of rats is increased after focal cerebral ischemia]. Sheng Li Xue Bao. 2004;56(2):153–7.

    CAS  PubMed  Google Scholar 

  131. Chinopoulos C et al. Inhibition of glutamate-induced delayed calcium deregulation by 2-APB and La3+ in cultured cortical neurones. J Neurochem. 2004;91(2):471–83.

    CAS  PubMed  Google Scholar 

  132. Marinelli S et al. Capsaicin activation of glutamatergic synaptic transmission in the rat locus coeruleus in vitro. J Physiol. 2002;543(Pt 2):531–40.

    CAS  PubMed  Google Scholar 

  133. Mezey É et al. Distribution of mRNA for vanilloid receptor subtype 1 (VR1), and VR1-like immunoreactivity, in the central nervous system of the rat and human. Proc Natl Acad Sci. 2000;97(7):3655–60.

    CAS  PubMed  Google Scholar 

  134. Hara Y et al. LTRPC2 Ca2+-permeable channel activated by changes in redox status confers susceptibility to cell death. Mol Cell. 2002;9(1):163–73.

    CAS  PubMed  Google Scholar 

  135. Miller BA. Inhibition of TRPM2 function by PARP inhibitors protects cells from oxidative stress-induced death. Br J Pharmacol. 2004;143(5):515–6.

    CAS  PubMed  Google Scholar 

  136. Miller BA. The role of TRP channels in oxidative stress-induced cell death. J Membr Biol. 2006;209(1):31–41.

    CAS  PubMed  Google Scholar 

  137. Pegorini S et al. Capsaicin exhibits neuroprotective effects in a model of transient global cerebral ischemia in Mongolian gerbils. Br J Pharmacol. 2005;144(5):727–35.

    CAS  PubMed  Google Scholar 

  138. Pegorini S et al. Vanilloid VR1 receptor is involved in rimonabant-induced neuroprotection. Br J Pharmacol. 2006;147(5):552–9.

    CAS  PubMed  Google Scholar 

  139. Berger C et al. Massive accumulation of N-acylethanolamines after stroke. Cell signalling in acute cerebral ischemia? J Neurochem. 2004;88(5):1159–67.

    CAS  PubMed  Google Scholar 

  140. Aarts M, Tymianski M. TRPMs and neuronal cell death. Pflugers Arch. 2005;451(1):243–9.

    CAS  PubMed  Google Scholar 

  141. Aarts M et al. A key role for TRPM7 channels in anoxic neuronal death. Cell. 2003;115(7):863–77.

    CAS  PubMed  Google Scholar 

  142. Contreras JE et al. Role of connexin-based gap junction channels and hemichannels in ischemia-induced cell death in nervous tissue. Brain Res Rev. 2004;47(1–3):290–303.

    CAS  PubMed Central  PubMed  Google Scholar 

  143. Contreras JE et al. Metabolic inhibition induces opening of unapposed connexin 43 gap junction hemichannels and reduces gap junctional communication in cortical astrocytes in culture. Proc Natl Acad Sci. 2002;99(1):495–500.

    CAS  PubMed  Google Scholar 

  144. Retamal MA et al. S-nitrosylation and permeation through connexin 43 hemichannels in astrocytes: induction by oxidant stress and reversal by reducing agents. Proc Natl Acad Sci U S A. 2006;103(12):4475–80.

    CAS  PubMed Central  PubMed  Google Scholar 

  145. Thompson RJ, Zhou N, MacVicar BA. Ischemia opens neuronal gap junction hemichannels. Science. 2006;312(5775):924–7.

    CAS  PubMed  Google Scholar 

  146. Farahani R et al. Alterations in metabolism and gap junction expression may determine the role of astrocytes as “good Samaritans” or executioners. Glia. 2005;50(4):351–61.

    PubMed  Google Scholar 

  147. Nakase T et al. Increased apoptosis and inflammation after focal brain ischemia in mice lacking connexin43 in astrocytes. Am J Pathol. 2004;164(6):2067–75.

    CAS  PubMed  Google Scholar 

  148. Oguro K et al. Global ischemia-induced increases in the gap junctional proteins connexin 32 (Cx32) and Cx36 in hippocampus and enhanced vulnerability of Cx32 knock-out mice. J Neurosci. 2001;21(19):7534–42.

    CAS  PubMed  Google Scholar 

  149. Nakase T, Yoshida Y, Nagata K. Enhanced connexin 43 immunoreactivity in penumbral areas in the human brain following ischemia. Glia. 2006;54(5):369–75.

    PubMed  Google Scholar 

  150. Lin JHC et al. Gap-junction-mediated propagation and amplification of cell injury. Nat Neurosci. 1998;1(6):494–500.

    CAS  PubMed  Google Scholar 

  151. Frantseva MV, Kokarovtseva L, Perez Velazquez JL. Ischemia-induced brain damage depends on specific gap-junctional coupling. J Cereb Blood Flow Metab. 2002;22(4):453–62.

    PubMed  Google Scholar 

  152. de Pina-Benabou MH et al. Blockade of gap junctions in vivo provides neuroprotection after perinatal global ischemia. Stroke. 2005;36(10):2232–7.

    PubMed  Google Scholar 

  153. Eggermont J et al. Cellular function and control of volume-regulated anion channels. Cell Biochem Biophys. 2001;35(3):263–74.

    CAS  PubMed  Google Scholar 

  154. Barakat AI et al. A flow-activated chloride-selective membrane current in vascular endothelial cells. Circ Res. 1999;85(9):820–8.

    CAS  PubMed  Google Scholar 

  155. Shen MR et al. Differential expression of volume-regulated anion channels during cell cycle progression of human cervical cancer cells. J Physiol. 2000;529(Pt 2):385–94.

    CAS  PubMed  Google Scholar 

  156. Okada Y et al. Receptor-mediated control of regulatory volume decrease (RVD) and apoptotic volume decrease (AVD). J Physiol. 2001;532(Pt 1):3–16.

    CAS  PubMed  Google Scholar 

  157. Jackson PS, Morrison R, Strange K. The volume-sensitive organic osmolyte-anion channel VSOAC is regulated by nonhydrolytic ATP binding. Am J Physiol - Cell Physiol. 1994;267(5):C1203–9.

    CAS  Google Scholar 

  158. Kimelberg HK. Increased release of excitatory amino acids by the actions of ATP and peroxynitrite on volume-regulated anion channels (VRACs) in astrocytes. Neurochem Int. 2004;45(4):511–9.

    CAS  PubMed  Google Scholar 

  159. Feustel PJ, Jin Y, Kimelberg HK. Volume-regulated anion channels are the predominant contributors to release of excitatory amino acids in the ischemic cortical penumbra. Stroke. 2004;35(5):1164–8.

    CAS  PubMed  Google Scholar 

  160. Kimelberg HK. Astrocytic swelling in cerebral ischemia as a possible cause of injury and target for therapy. Glia. 2005;50(4):389–97.

    PubMed  Google Scholar 

  161. Patel AJ et al. Disruption of mitochondrial respiration inhibits volume-regulated anion channels and provokes neuronal cell swelling. J Neurosci. 1998;18(9):3117–23.

    CAS  PubMed  Google Scholar 

  162. Zhang Y et al. DCPIB, a specific inhibitor of volume regulated anion channels (VRACs), reduces infarct size in MCAo and the release of glutamate in the ischemic cortical penumbra. Exp Neurol. 2008;210(2):514–20.

    CAS  PubMed Central  PubMed  Google Scholar 

  163. Seki Y et al. Inhibition of ischemia-induced glutamate release in rat striatum by dihydrokinate and an anion channel blocker. Stroke. 1999;30(2):433–40.

    CAS  PubMed  Google Scholar 

  164. Liu H-T et al. Roles of two types of anion channels in glutamate release from mouse astrocytes under ischemic or osmotic stress. Glia. 2006;54(5):343–57.

    PubMed  Google Scholar 

  165. Yamamoto N et al. Alterations in the expression of the AQP family in cultured rat astrocytes during hypoxia and reoxygenation. Mol Brain Res. 2001;90(1):26–38.

    CAS  PubMed  Google Scholar 

  166. Taniguchi M et al. Induction of aquaporin-4 water channel mRNA after focal cerebral ischemia in rat. Mol Brain Res. 2000;78(1–2):131–7.

    CAS  PubMed  Google Scholar 

  167. Manley GT et al. Aquaporin-4 deletion in mice reduces brain edema after acute water intoxication and ischemic stroke. Nat Med. 2000;6(2):159–63.

    CAS  PubMed  Google Scholar 

  168. Papadopoulos MC, Manley GT, Krishna Sanjeev, Verkman AS. “Aquaporin-4 facilitates reabsorption of excess fluid in vasogenic brain edema.” FASEB J. 2004;18(11):1291–93.

    Google Scholar 

  169. Huber VJ et al. Identification of arylsulfonamides as aquaporin 4 inhibitors. Bioorg Med Chem Lett. 2007;17(5):1270–3.

    CAS  PubMed  Google Scholar 

  170. Detmers FJM et al. Quaternary ammonium compounds as water channel blockers: specificity, potency, and site of action. J Biol Chem. 2006;281(20):14207–14.

    CAS  PubMed  Google Scholar 

  171. Niemietz CM, Tyerman SD. New potent inhibitors of aquaporins: silver and gold compounds inhibit aquaporins of plant and human origin. FEBS Lett. 2002;531(3):443–7.

    CAS  PubMed  Google Scholar 

  172. Ito H et al. Interleukin-1β induces the expression of aquaporin-4 through a nuclear factor-κB pathway in rat astrocytes. J Neurochem. 2006;99(1):107–18.

    CAS  PubMed  Google Scholar 

  173. Higashida T et al. The role of hypoxia-inducible factor-1α, aquaporin-4, and matrix metalloproteinase-9 in blood–brain barrier disruption and brain edema after traumatic brain injury. J Neurosurg. 2011;114(1):92–101.

    CAS  PubMed  Google Scholar 

Download references

Acknowledgment

This work was supported by grants to JMS from the National Heart, Lung and Blood Institute (HL082517) and the National Institute of Neurological Disorders and Stroke (NS061808) and to BPW from the Brain Aneurysm Foundation.

Compliance with Ethics Requirements

JMS holds a US patent (#7, 285, 574), “A novel non-selective cation channel in neural cells and methods for treating brain swelling,” and is a member of the Scientific Advisory Board and holds shares in Remedy Pharmaceuticals. No support was provided by Remedy Pharmaceuticals to JMS for this project.

This article does not contain any studies with human or animal subjects.

Conflict of Interest

Arjun Khanna, Kristopher T. Kahle, Brian P. Walcott, Volodymyr Gerzanich, and J Marc Simard declare that they have no conflict of interest.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Brian P. Walcott or J. Marc Simard.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Khanna, A., Kahle, K.T., Walcott, B.P. et al. Disruption of Ion Homeostasis in the Neurogliovascular Unit Underlies the Pathogenesis of Ischemic Cerebral Edema. Transl. Stroke Res. 5, 3–16 (2014). https://doi.org/10.1007/s12975-013-0307-9

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12975-013-0307-9

Keywords

Navigation