Skip to main content
Log in

The Potential Therapeutic Capacity of Inhibiting the Brain Renin–Angiotensin System in the Treatment of Co-Morbid Conditions in Epilepsy

  • Review Article
  • Published:
CNS Drugs Aims and scope Submit manuscript

Abstract

Epilepsy is one of the most prevalent neurological diseases and although numerous novel anticonvulsants have been approved, the proportion of patients who are refractory to medical treatment of seizures and have progressive co-morbidities such as cognitive impairment and depression remains at about 20–30%. In the last decade, extensive research has identified a therapeutic capacity of the components of the brain renin–angiotensin system (RAS) in seizure- and epilepsy-related phenomena. Alleviating the activity of RAS in the central nervous system is considered to be a potential adjuvant strategy for the treatment of numerous detrimental consequences of epileptogenesis. One of the main advantages of RAS is associated with its modulatory influence on different neurotransmitter systems, thereby exerting a fine-tuning control mechanism for brain excitability. The most recent scientific findings regarding the involvement of the components of brain RAS show that angiotensin II (Ang II), angiotensin-converting enzyme (ACE), Ang II type 1 (AT1) and type 2 (AT2) receptors are involved in the control of epilepsy and its accompanying complications, and therefore they are currently of therapeutic interest in the treatment of this disease. However, data on the role of different components of brain RAS on co-morbid conditions in epilepsy, including hypertension, are insufficient. Experimental and clinical findings related to the involvement of Ang II, ACE, AT1, and AT2 receptors in the control of epilepsy and accompanying complications may point to new therapeutic opportunities and adjuvants for the treatment of common co-morbid conditions of epilepsy.

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.

Institutional subscriptions

Fig. 1

Similar content being viewed by others

References

  1. Quigg M, Straume M, Smith T, Menaker M, Bertram EH. Seizures induce phase shifts of rat circadian rhythms. Brain Res. 2001;913:165–9. https://doi.org/10.1016/S0006-8993(01)02780-9.

    Article  CAS  PubMed  Google Scholar 

  2. Shin HW, Davis R. Review of levetiracetam as a first line treatment in status epilepticus in the adult patients—what do we know so far? Front Neurol. 2013;5(4):111. https://doi.org/10.3389/fneur.2013.00111.

    Article  Google Scholar 

  3. Tchekalarova J, Pechlivanova D, Atanasova T, Markova P, Lozanov V, Stoynev A. Diurnal variations in depression-like behavior of Wistar and spontaneously hypertensive rats in the kainate model of temporal lobe epilepsy. Epilepsy Behav. 2011;20:277–85. https://doi.org/10.1016/j.yebeh.2010.12.021.

    Article  PubMed  Google Scholar 

  4. Watanabe M, Forsgren L, Tomson T, Mathern GW, Glynn M, Engel J, et al. ILAE official report: a practical clinical definition of epilepsy. Brain Res. 2014;55:475–82. https://doi.org/10.1111/epi.12550.

    Article  Google Scholar 

  5. Sloviter RS. Apoptosis: a guide for the perplexed. Trends Pharmacol Sci. 2002;23:19–24. https://doi.org/10.1016/S0165-6147(00)01867-8.

    Article  CAS  PubMed  Google Scholar 

  6. Dobolyi A, Kékesi KA, Juhász G, Székely AD, Lovas G, Kovács Z. Receptors of peptides as therapeutic targets in epilepsy research. Curr Med Chem. 2014;21:764–87.

    Article  CAS  Google Scholar 

  7. Kanner AM, Balabanov A. Valproate: a practical review of its uses in neurological and psychiatric disorders. Expert Rev Neurother. 2002;2:151–65. https://doi.org/10.1586/14737175.2.2.151.

    Article  CAS  PubMed  Google Scholar 

  8. Gaitatzis A, Carroll K, Majeed A, Sander JW. The epidemiology of the comorbidity of epilepsy in the general population. Brain Res. 2004;45:1613–22. https://doi.org/10.1111/j.0013-9580.2004.17504.x.

    Article  Google Scholar 

  9. Lacey CJ, Salzberg MR, Roberts H, Trauer T, D’Souza WJ. Psychiatric comorbidity and impact on health service utilization in a community sample of patients with epilepsy. Brain Res. 2009;50:1991–4. https://doi.org/10.1111/j.1528-1167.2009.02165.x.

    Article  Google Scholar 

  10. Kovac S, Walker MC. Neuropeptides in epilepsy. Neuropeptides. 2013;47:467–75. https://doi.org/10.1016/j.npep.2013.10.015.

    Article  CAS  PubMed  Google Scholar 

  11. Atanasova M, Petkova Z, Pechlivanova D, Dragomirova P, Blazhev A, Tchekalarova J. Strain-dependent effects of long-term treatment with melatonin on kainic acid-induced status epilepticus, oxidative stress and the expression of heat shock proteins. Pharmacol Biochem Behav. 2013;111:44–50. https://doi.org/10.1016/j.pbb.2013.08.006.

    Article  CAS  PubMed  Google Scholar 

  12. Wright JW, Harding JW. Brain renin-angiotensin—a new look at an old system. Prog Neurobiol. 2011;95:49–67. https://doi.org/10.1016/j.pneurobio.2011.07.001.

    Article  CAS  PubMed  Google Scholar 

  13. Wright JW, Harding JW. The brain renin-angiotensin system: a diversity of functions and implications for CNS diseases. Pflugers Arch. 2013;465:133–51. https://doi.org/10.1007/s00424-012-1102-2.

    Article  CAS  PubMed  Google Scholar 

  14. De Bundel D, Smolders I, Vanderheyden P, Michotte Y. Ang II and Ang IV: unraveling the mechanism of action on synaptic plasticity, memory, and epilepsy. CNS Neurosci Ther. 2008;14:315–39. https://doi.org/10.1111/j.1755-5949.2008.00057.x.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Stragier B, De Bundel D, Sarre S, Smolders I, Vauquelin G, Dupont A, et al. Involvement of insulin-regulated aminopeptidase in the effects of the renin–angiotensin fragment angiotensin IV: a review. Heart Fail Rev. 2008;13:321–37. https://doi.org/10.1007/s10741-007-9062-x.

    Article  CAS  PubMed  Google Scholar 

  16. Dzau VJ, Bernstein K, Celermajer D, Cohen J, Dahlöf B, Deanfield J, et al. The relevance of tissue angiotensin-converting enzyme: manifestations in mechanistic and endpoint data. Am J Cardiol. 2001;88:1–20. https://doi.org/10.1016/S0002-9149(01)01878-1.

    Article  Google Scholar 

  17. Kaschina E, Unger T. Angiotensin AT1/AT2 receptors: regulation, signalling and function. Blood Press. 2003;12:70–88.

    Article  CAS  Google Scholar 

  18. Carey RM. Update on the role of the AT2 receptor. Curr Opin Nephrol Hypertens. 2005;14:67–71. https://doi.org/10.1097/00041552-200501000-00011.

    Article  CAS  PubMed  Google Scholar 

  19. Argañaraz GA, Konno AC, Perosa SR, Santiago JFC, Boim MA, Vidotti DB, et al. The renin-angiotensin system is upregulated in the cortex and hippocampus of patients with temporal lobe epilepsy related to mesial temporal sclerosis. Brain Res. 2008;49:1348–57. https://doi.org/10.1111/j.1528-1167.2008.01581.x.

    Article  CAS  Google Scholar 

  20. Pereira MGAG, Becari C, Oliveira JAC, Salgado MCO, Garcia-Cairasco N, Costa-Neto CM. Inhibition of the renin–angiotensin system prevents seizures in a rat model of epilepsy. Clin Sci. 2010;119:477–82. https://doi.org/10.1042/CS20100053.

    Article  CAS  PubMed  Google Scholar 

  21. Gouveia TLF, Frangiotti MIB, De Brito JMV, De Castro Neto EF, Sakata MM, Febba AC, et al. The levels of renin-angiotensin related components are modified in the hippocampus of rats submitted to pilocarpine model of epilepsy. Neurochem Int. 2012;61:54–62. https://doi.org/10.1016/j.neuint.2012.04.012.

    Article  CAS  PubMed  Google Scholar 

  22. Atanasova D, Tchekalarova J, Ivanova N, Nenchovska Z, Pavlova E, Atanassova N, et al. Losartan suppresses the kainate-induced changes of angiotensin AT1receptor expression in a model of comorbid hypertension and epilepsy. Life Sci. 2018;193:40–6. https://doi.org/10.1016/j.lfs.2017.12.006.

    Article  CAS  PubMed  Google Scholar 

  23. Bar-Klein G, Cacheaux LP, Kamintsky L, Prager O, Weissberg I, Schoknecht K, et al. Losartan prevents acquired epilepsy via TGF-β signaling suppression. Ann Neurol. 2014;75:864–75. https://doi.org/10.1002/ana.24147.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Sun H, Wu HQ, Yu X, Zhang GL, Zhang R, Zhan SQ, et al. Angiotensin II and its receptor in activated microglia enhanced neuronal loss and cognitive impairment following pilocarpine-induced status epilepticus. Mol Cell Neurosci. 2015;65:58–67. https://doi.org/10.1016/j.mcn.2015.02.014.

    Article  CAS  PubMed  Google Scholar 

  25. Tchekalarova JD, Ivanova NM, Pechlivanova DM, Atanasova D, Lazarov N, Kortenska L, et al. Antiepileptogenic and neuroprotective effects of losartan in kainate model of temporal lobe epilepsy. Pharmacol Biochem Behav. 2014;127:27–36. https://doi.org/10.1016/j.pbb.2014.10.005.

    Article  CAS  PubMed  Google Scholar 

  26. Tchekalarova JD, Ivanova N, Atanasova D, Pechlivanova DM, Lazarov N, Kortenska L, et al. Long-term treatment with losartan attenuates seizure activity and neuronal damage without affecting behavioral changes in a model of co-morbid hypertension and epilepsy. Cell Mol Neurobiol. 2016;36:927–41. https://doi.org/10.1007/s10571-015-0278-3.

    Article  CAS  PubMed  Google Scholar 

  27. Tchekalarova J, Kambourova T, Georgiev V. Long-term theophylline treatment changes the effects of angiotensin II and adenosinergic agents on the seizure threshold. Brain Res Bull. 2000;52:13–6. https://doi.org/10.1016/S0361-9230(99)00254-3.

    Article  CAS  PubMed  Google Scholar 

  28. Tchekalarova J, Georgiev V. Ang II and Ang III modulate PTZ seizure threshold in non-stressed and stressed mice: possible involvement of noradrenergic mechanism. Neuropeptides. 2006;40:339–48. https://doi.org/10.1016/j.npep.2006.07.005.

    Article  CAS  PubMed  Google Scholar 

  29. Tchekalarova J, Georgiev V. Adenosine-angiotensin II interactions in pentylenetetrazol seizure threshold in mice. J Physiol Paris. 1999;93:191–7. https://doi.org/10.1016/S0928-4257(99)80151-X.

    Article  CAS  PubMed  Google Scholar 

  30. Tchekalarova J, Georgiev V. Angiotensin peptides modulatory system: how is it implicated in the control of seizure susceptibility? Life Sci. 2005;76:955–70. https://doi.org/10.1016/j.lfs.2004.10.012.

    Article  CAS  PubMed  Google Scholar 

  31. Tchekalarova J, Kambourova T, Georgiev V. Effects of angiotensin III and angiotensin IV on pentylenetetrazol seizure susceptibility (threshold and kindling): interaction with adenosine A1 receptors. Brain Res Bull. 2001;56:87–91. https://doi.org/10.1016/S0361-9230(01)00568-8.

    Article  CAS  PubMed  Google Scholar 

  32. Ivanova NM, Atanasova D, Pechlivanova DM, Mitreva R, Lazarov N, Stoynev AG, et al. Long-term intracerebroventricular infusion of angiotensin II after kainate-induced status epilepticus: effects on epileptogenesis, brain damage, and diurnal behavioral changes. Epilepsy Behav. 2015;51:1–12. https://doi.org/10.1016/j.yebeh.2015.06.036.

    Article  PubMed  Google Scholar 

  33. Esclapez M, Hirsch JC, Ben-Ari Y, Bernard C. Newly formed excitatory pathways provide a substrate for hyperexcitability in experimental temporal lobe epilepsy. J Comp Neurol. 1999;408:449–60. https://doi.org/10.1002/(SICI)1096-9861(19990614)408:4%3c449:AID-CNE1%3e3.0.CO;2-R.

    Article  CAS  PubMed  Google Scholar 

  34. Tsunashima K, Schwarzer C, Kirchmair E, Sieghart W, Sperk G. GABAA receptor subunits in the rat hippocampus III: altered messenger RNA expression in kainic acid-induced epilepsy. Neuroscience. 1997;80:1019–32. https://doi.org/10.1016/S0306-4522(97)00144-9.

    Article  CAS  PubMed  Google Scholar 

  35. Sloviter RS. Decreased hippocampal inhibition and a selective loss of interneurons in experimental epilepsy. Science. 1987;235:73–6. https://doi.org/10.1126/science.2879352.

    Article  CAS  PubMed  Google Scholar 

  36. Tauck DL, Nadler JV. Evidence of functional mossy fiber sprouting in hippocampal formation of kainic acid-treated rats. J Neurosci. 1985;5:1016–22. http://www.ncbi.nlm.nih.gov/pubmed/3981241.

    Article  CAS  Google Scholar 

  37. Klein P, Dingledine R, Aronica E, Bernard C, Blümcke I, Boison D, et al. Commonalities in epileptogenic processes from different acute brain insults: do they translate? Brain Res. 2018;59:37–66. https://doi.org/10.1111/epi.13965.

    Article  CAS  Google Scholar 

  38. Albrecht D, Broser M, Krüger H. Excitatory action of angiotensins II and IV on hippocampal neuronal activity in urethane anesthetized rats. Regul Pept. 1997;70:105–9. https://doi.org/10.1016/S0167-0115(97)00015-3.

    Article  CAS  PubMed  Google Scholar 

  39. Haas HL, Felix D, Celio MR, Inagami T. Angiotensin II in the hippocampus. A histochemical and electrophysiological study. Experientia. 1980;36:1394–5. https://doi.org/10.1007/BF01960117.

    Article  CAS  PubMed  Google Scholar 

  40. Latchford KJ, Ferguson AV. ANG II-induced excitation of paraventricular nucleus magnocellular neurons: a role for glutamate interneurons. Am J Physiol Regul Integr Comp Physiol. 2004;286:R894–902. https://doi.org/10.1152/ajpregu.00603.2003.

    Article  CAS  PubMed  Google Scholar 

  41. Albrecht D, Nitschke T, Von Bohlen und Halbach O. Various effects of angiotensin II on amygdaloid neuronal activity in normotensive control and hypertensive transgenic [TGR(mREN-2)27] rats. FASEB J. 2000;14:925–31. https://doi.org/10.1096/fasebj.14.7.925.

    Article  CAS  PubMed  Google Scholar 

  42. Łukawski K, Janowska A, Jakubus T, Tochman-Gawda A, Czuczwar SJ. Angiotensin AT1 receptor antagonists enhance the anticonvulsant action of valproate in the mouse model of maximal electroshock. Eur J Pharmacol. 2010;640:172–7. https://doi.org/10.1016/j.ejphar.2010.04.053.

    Article  CAS  PubMed  Google Scholar 

  43. Łukawski K, Janowska A, Jakubus T, Raszewski G, Czuczwar SJ. Combined treatment with gabapentin and drugs affecting the renin-angiotensin system against electroconvulsions in mice. Eur J Pharmacol. 2013. https://doi.org/10.1016/j.ejphar.2013.02.054.

    Article  PubMed  Google Scholar 

  44. Łukawski K, Janowska A, Jakubus T, Czuczwar SJ. Interactions between angiotensin AT1 receptor antagonists and second-generation antiepileptic drugs in the test of maximal electroshock. Fundam Clin Pharmacol. 2014;28:277–83. https://doi.org/10.1111/fcp.12023.

    Article  CAS  PubMed  Google Scholar 

  45. De Sarro G, Di Paola ED, Gratteri S, Gareri P, Rispoli V, Siniscalchi A, et al. Fosinopril and zofenopril, two angiotensin-converting enzyme (ACE) inhibitors, potentiate the anticonvulsant activity of antiepileptic drugs against audiogenic seizures in DBA/2 mice. Pharmacol Res. 2012;65:285–96. https://doi.org/10.1016/j.phrs.2011.11.005.

    Article  CAS  PubMed  Google Scholar 

  46. Łukawski K, Raszewski G, Czuczwar SJ. Effect of aliskiren, a direct renin inhibitor, on the protective action of antiepileptic drugs against pentylenetetrazole-induced clonic seizures in mice. Fundam Clin Pharmacol. 2019;33:191–8. https://doi.org/10.1111/fcp.12421.

    Article  CAS  PubMed  Google Scholar 

  47. Łukawski K, Raszewski G, Czuczwar SJ. Interactions of aliskiren, a direct renin inhibitor, with antiepileptic drugs in the test of maximal electroshock in mice. Eur J Pharmacol. 2018;819:108–13. https://doi.org/10.1016/j.ejphar.2017.11.037.

    Article  CAS  PubMed  Google Scholar 

  48. Russo E, Leo A, Scicchitano F, Donato A, Ferlazzo E, Gasparini S, et al. Cerebral small vessel disease predisposes to temporal lobe epilepsy in spontaneously hypertensive rats. Brain Res Bull. 2017;130:245–50. https://doi.org/10.1016/j.brainresbull.2017.02.003.

    Article  PubMed  Google Scholar 

  49. Pechlivanova DM, Stoynev AG, Tchekalarova JD. The effects of chronic losartan pretreatment on restraint stress-induced changes in motor activity, nociception and pentylenetetrazol generalized seizures in rats. Folia Med. 2011;53:69–73. https://doi.org/10.2478/v10153-010-0040-z.

    Article  Google Scholar 

  50. Pushpa VH. Evaluation and comparison of anticonvulsant activity of t elmisartan and olmesartan in experimentally induced animal models of epilepsy. J Clin Diagn Res. 2014;8:8–12. https://doi.org/10.7860/jcdr/2014/9455.5061.

    Article  Google Scholar 

  51. Tchekalarova J, Loyens E, Smolders I. Effects of AT1 receptor antagonism on kainate-induced seizures and concomitant changes in hippocampal extracellular noradrenaline, serotonin, and dopamine levels in Wistar-Kyoto and spontaneously hypertensive rats. Epilepsy Behav. 2015;46:66–71. https://doi.org/10.1016/j.yebeh.2015.03.021.

    Article  PubMed  Google Scholar 

  52. Wright JW, Yamamoto BJ, Harding JW. Angiotensin receptor subtype mediated physiologies and behaviors: new discoveries and clinical targets. Prog Neurobiol. 2008;84:157–81. https://doi.org/10.1016/j.pneurobio.2007.10.009.

    Article  CAS  PubMed  Google Scholar 

  53. Jöhren J, Saavedra O. Expression messenger of AT 1 ~ and ATlB angiotensin RNA in forebrain of 2-week-old 11 receptor rats. Am J Physiol. 1996;271:E104–12. https://doi.org/10.1152/ajpendo.1996.271.1.E104.

    Article  PubMed  Google Scholar 

  54. Chen QH, Toney GM. Responses to GABA-A receptor blockade in the hypothalamic PVN are attenuated by local AT 1 receptor antagonism. Am J Physiol Regul Integr Comp Physiol. 2003;85:R1231–9. https://doi.org/10.1152/ajpregu.00028.2003.

    Article  Google Scholar 

  55. Unger T, Bles F, Ganten D, Lang RE, Rettig R, Schwab NA. Gabaergic stimulation inhibits central actions of angiotensin II: pressor responses, drinking and release of vasopressin. Eur J Pharmacol. 1983;90:1–9. https://doi.org/10.1016/0014-2999(83)90207-8.

    Article  CAS  PubMed  Google Scholar 

  56. Brandt C, Gastens AM, Sun M, Hausknecht M, Löscher W. Treatment with valproate after status epilepticus: effect on neuronal damage, epileptogenesis, and behavioral alterations in rats. Neuropharmacology. 2006;51:789–804. https://doi.org/10.1016/j.neuropharm.2006.05.021.

    Article  CAS  PubMed  Google Scholar 

  57. Detour J, Schroeder H, Desor D, Nehlig A. A 5-month period of epilepsy impairs spatial memory, decreases anxiety, but spares object recognition in the lithium–pilocarpine model in adult rats. Brain Res. 2005;46:499–508. https://doi.org/10.1111/j.0013-9580.2005.38704.x.

    Article  Google Scholar 

  58. Stafstrom CE, Chronopoulos A, Thurber S, Thompson JL, Holmes GL. Age-dependent cognitive and behavioral deficits after kainic acid seizures. Brain Res. 1993;34:420–32. https://doi.org/10.1111/j.1528-1157.1993.tb02582.x.

    Article  CAS  Google Scholar 

  59. Villapol S, Yaszemski AK, Logan TT, Sánchez-Lemus E, Saavedra JM, Symes AJ. Candesartan, an angiotensin II at 1-receptor blocker and PPAR-γ agonist, reduces lesion volume and improves motor and memory function after traumatic brain injury in mice. Neuropsychopharmacology. 2012;37:2817–29. https://doi.org/10.1038/npp.2012.152.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Nayak V, Patil PA. Antidepressant activity of fosinopril, ramipril and losartan, but not of lisinopril in depressive paradigms of albino rats and mice. Ind J Exp Biol. 2008;46:180–4.

    CAS  Google Scholar 

  61. Saavedra JM, Ando H, Armando I, Baiardi G, Bregonzio C, Juorio A, et al. Anti-stress and anti-anxiety effects of centrally acting angiotensin II AT1 receptor antagonists. Regul Pept. 2005;128:227–38. https://doi.org/10.1016/j.regpep.2004.12.015.

    Article  CAS  PubMed  Google Scholar 

  62. Saavedra JM, Benicky J, Zhou J. Angiotensin II: multitasking in the brain. J Hypertens. 2006;24:131–7. https://doi.org/10.1097/01.hjh.0000220418.09021.ee.

    Article  CAS  Google Scholar 

  63. Saavedra JM, Sánchez-Lemus E, Benicky J. Blockade of brain angiotensin II AT1 receptors ameliorates stress, anxiety, brain inflammation and ischemia: therapeutic implications. Psychoneuroendocrinology. 2011;36:1–18. https://doi.org/10.1016/j.psyneuen.2010.10.001.

    Article  CAS  PubMed  Google Scholar 

  64. de los Marinzalda MA, Bregonzio C, Baiardi G, Casarsa BS, Gargiulo PA, Pérez PA. Fear-potentiated behaviour is modulated by central amygdala angiotensin II AT1 receptors stimulation. Biomed Res Int. 2014;2014:183248. https://doi.org/10.1155/2014/183248.

    Article  CAS  PubMed Central  Google Scholar 

  65. Giardina WJ, Ebert DM. Positive effects of captopril in the behavioral despair swim test. Biol Psychiatry. 1989;25:697–702. https://doi.org/10.1016/0006-3223(89)90240-0.

    Article  CAS  PubMed  Google Scholar 

  66. Ayyub M, Najmi AK, Akhtar M. Protective effect of irbesartan an angiotensin (AT 1) receptor antagonist in unpredictable chronic mild stress induced depression in mice. Drug Res (Stuttg). 2017;67:59–64. https://doi.org/10.1055/s-0042-118172.

    Article  CAS  PubMed  Google Scholar 

  67. Ping G, Qian W, Song G, Zhaochun S. Valsartan reverses depressive/anxiety-like behavior and induces hippocampal neurogenesis and expression of BDNF protein in unpredictable chronic mild stress mice. Pharmacol Biochem Behav. 2014;124:5–12. https://doi.org/10.1016/j.pbb.2014.05.006.

    Article  CAS  PubMed  Google Scholar 

  68. Martin P, Massol J, Puech AJ. Captopril as an antidepressant? Effects on the learned helplessness paradigm in rats. Biol Psychiatry. 1990;27:968–74. https://doi.org/10.1016/0006-3223(90)90034-Y.

    Article  CAS  PubMed  Google Scholar 

  69. Aswar U, Chepurwar S, Shintre S, Aswar M. Telmisartan attenuates diabetes induced depression in rats. Pharmacol Rep. 2017;69:358–64. https://doi.org/10.1016/j.pharep.2016.12.004.

    Article  CAS  PubMed  Google Scholar 

  70. Chalas A, Conway EL. No evidence for involvement of angiotensin II in spatial learning in water maze in rats. Behav Brain Res. 1996;81:199–205. https://doi.org/10.1016/S0166-4328(96)00062-9.

    Article  CAS  PubMed  Google Scholar 

  71. Tota S, Goel R, Pachauri SD, Najmi AK, Hanif K, Nath C. Effect of angiotensin II on spatial memory, cerebral blood flow, cholinergic neurotransmission, and brain derived neurotrophic factor in rats. Psychopharmacology. 2013;226:357–69. https://doi.org/10.1007/s00213-012-2913-8.

    Article  CAS  PubMed  Google Scholar 

  72. Mogi M, Iwanami J, Horiuchi M. Roles of brain angiotensin II in cognitive function and dementia. Int J Hypertens. 2012;2012:169649. https://doi.org/10.1155/2012/169649.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Ivanova N, Tchekalarova J, Atanasova D, Pechlivanova D, Lazarov N. Strain-dependent effects of AT1 receptor antagonist losartan on spatial memory performance of wistar and spontaneously hypertensive rats in kainate model of temporal epilepsy. Epilepsy Behav. 2018;71:839–46. https://doi.org/10.7546/CRABS.2018.06.15.

    Article  CAS  Google Scholar 

  74. Herrero AI, Sandi C, Venero C. Individual differences in anxiety trait are related to spatial learning abilities and hippocampal expression of mineralocorticoid receptors. Neurobiol Learn Mem. 2006;86:150–9. https://doi.org/10.1016/j.nlm.2006.02.001.

    Article  CAS  PubMed  Google Scholar 

  75. Schmidt R, Payer F, Niederkorn K, Offenbacher H, Horner S, Blematl B, et al. Magnetic resonance imaging white matter lesions and cognitive impairment in hypertensive individuals. Arch Neurol. 1991;48:417–20. https://doi.org/10.1001/archneur.1991.00530160087019.

    Article  CAS  PubMed  Google Scholar 

  76. Wyss JM, Kadish I, Van Groen T. Age-related decline in spatial learning and memory: attenuation by captopril. Clin Exp Hypertens. 2003;7:455–74. https://doi.org/10.1081/CEH-120024988.

    Article  CAS  Google Scholar 

  77. Grammatopoulos TN, Ahmadi F, Jones SM, Fariss MW, Weyhenmeyer JA, Zawada WM. Angiotensin II protects cultured midbrain dopaminergic neurons against rotenone-induced cell death. Brain Res. 2005;1045:64–71. https://doi.org/10.1016/j.brainres.2005.03.038.

    Article  CAS  PubMed  Google Scholar 

  78. Wilms H, Rosenstiel P, Unger T, Deuschl G, Lucius R. Neuroprotection with angiotensin receptor antagonists: a review of the evidence and potential mechanisms. Am J Cardiovasc Drugs. 2005;5:245–53. https://doi.org/10.2165/00129784-200505040-00004.

    Article  CAS  PubMed  Google Scholar 

  79. Armstrong DL, Garcia EA, Ma T, Quinones B, Wayner MJ. Angiotensin II blockade of long-term potentiation at the perforant path-granule cell synapse in vitro. Peptides. 1996;17:689–93. https://doi.org/10.1016/0196-9781(96)00030-7.

    Article  CAS  PubMed  Google Scholar 

  80. Almeida SS, Naffah-Mazzacoratti MG, Guimarães PB, Wasinski F, Pereira FE, Canzian M, et al. Carbamazepine inhibits angiotensin I-converting enzyme, linking it to the pathogenesis of temporal lobe epilepsy. Transl Psychiatry. 2012;2:93. https://doi.org/10.1038/tp.2012.21.

    Article  CAS  Google Scholar 

  81. Reyes-Garcia SZ, Scorza CA, Ortiz-Villatoro NN, Cavalheiro EA. Losartan fails to suppress epileptiform activity in brain slices from resected tissues of patients with drug resistant epilepsy. J Neurol Sci. 2019;397:169–71. https://doi.org/10.1016/j.jns.2019.01.008.

    Article  CAS  PubMed  Google Scholar 

  82. Delanty NVC. Seizures, hypertension, and posterior leukoencephalopathy. New York: Springer Science; 2002. p. 251–60.

    Google Scholar 

  83. Gasparini S, Ferlazzo E, Sueri C, Cianci V, Ascoli M, Cavalli SM, Epilepsy Study Group of the Italian Neurological Society. Hypertension, seizures, and epilepsy: a review on pathophysiology and management. Neurol Sci. 2019;40(9):1775–83. https://doi.org/10.1007/s10072-019-03913-4.

    Article  PubMed  Google Scholar 

  84. Tchekalarova J, Ivanova N, Pechlivanova D, Ilieva K, Atanasova M. Strain-dependent effects of sub-chronically infused losartan against kainic acid-induced seizures, oxidative stress, and heat shock protein 72 expression. Cell Mol Neurobiol. 2014;34:133–42. https://doi.org/10.1007/s10571-013-9994-8.

    Article  CAS  PubMed  Google Scholar 

  85. Vorobyov V, Schibaev N, Kaptsov V, Kovalev G, Sengpiel F. Cortical and hippocampal EEG effects of neurotransmitter agonists in spontaneously hypertensive vs. kainate-treated rats. Brain Res. 2011;1383:154–68. https://doi.org/10.1016/j.brainres.2011.01.107.

    Article  CAS  PubMed  Google Scholar 

  86. Romero-Nava R, Rodriguez JE, Reséndiz-Albor AA, Sánchez-Munõz F, Ruiz-Hernandéz A, Huang F, et al. Changes in protein and gene expression of angiotensin II receptors (AT1 and AT2) in aorta of diabetic and hypertensive rats. Clin Exp Hypertens. 2016;38:56–62. https://doi.org/10.3109/10641963.2015.1060984.

    Article  CAS  PubMed  Google Scholar 

  87. Johansson B. Indomethacin and cerebrovascular permeability to albumin in acute hypertension and cerebral embolism in the rat. Exp Brain Res. 1980;42:331–6.

    Google Scholar 

  88. Ndode-Ekane XE, Hayward N, Gröhn O, Pitkänen A. Vascular changes in epilepsy: functional consequences and association with network plasticity in pilocarpine-induced experimental epilepsy. Neuroscience. 2010;166:312–32. https://doi.org/10.1016/j.neuroscience.2009.12.002.

    Article  CAS  PubMed  Google Scholar 

  89. Cornford EM, Oldendorf W. Epilepsy and the blood–brain barrier. Adv Neurol. 1986;44:787–812.

    CAS  PubMed  Google Scholar 

  90. Gorter JA, Van Vliet EA, Aronica E. Status epilepticus, blood–brain barrier disruption, inflammation, and epileptogenesis. Epilepsy Behav. 2015;49:13–6. https://doi.org/10.1016/j.yebeh.2015.04.047.

    Article  PubMed  Google Scholar 

  91. Oztaş B, Kaya M. The effect of acute hypertension on blood–brain barrier permeability to albumin during experimentally induced epileptic seizures. Pharmacol Res. 1991;23(1):41–6. https://doi.org/10.1016/s1043-6618(05)80104-5.

    Article  PubMed  Google Scholar 

  92. Kucuk M, Kaya M, Kalayci R, Cimen V, Kudat H, Arican N, et al. Effects of losartan on the blood–brain barrier permeability in long-term nitric oxide blockade-induced hypertensive rats. Life Sci. 2002;71:937–46. https://doi.org/10.1016/S0024-3205(02)01772-1.

    Article  CAS  PubMed  Google Scholar 

  93. Kaya M, Kalayci R, Küçük M, Arican N, Elmas I, Kudat H, et al. Effect of losartan on the blood–brain barrier permeability in diabetic hypertensive rats. Life Sci. 2003;73:3235–44. https://doi.org/10.1016/j.lfs.2003.06.014.

    Article  CAS  PubMed  Google Scholar 

  94. Ye S, Zhong H, Duong VN, Campese VM. Losartan reduces central and peripheral sympathetic nerve activity in a rat model of neurogenic hypertension. Hypertension. 2002;39:1101–6. https://doi.org/10.1161/01.HYP.0000018590.26853.C7.

    Article  CAS  PubMed  Google Scholar 

  95. Gasparini S, Ferlazzo E, Beghi E, Sofia V, Mumoli L, Labate A. Epilepsy associated with leukoaraiosis mainly affects temporal lobe: a casual or causal relationship? Epilepsy Res. 2015;109:1–8. https://doi.org/10.1016/j.eplepsyres.2014.10.012.

    Article  PubMed  Google Scholar 

  96. Ferlazzo E, Gasparini S, Beghi E, Sueri C, Russo E, Leo A. Epilepsy in cerebrovascular diseases: review of experimental and clinical data with meta-analysis of risk factors. Epilepsia. 2016;57(8):1205–14. https://doi.org/10.1111/epi.13448.

    Article  PubMed  Google Scholar 

  97. Haspula D, Clark MA. Neuroinflammation and sympathetic overactivity: mechanisms and implications in hypertension. Auton Neurosci. 2018;210:10–7. https://doi.org/10.1016/j.autneu.2018.01.002.

    Article  CAS  PubMed  Google Scholar 

  98. Pietranera L, Saravia F, Gonzalez Deniselle MC, Roig P, Lima A, De Nicola AF. Abnormalities of the hippocampus are similar in deoxycorticosterone acetate-salt hypertensive rats and spontaneously hypertensive rats. J Neuroendocrinol. 2006;18:466–74. https://doi.org/10.1111/j.1365-2826.2006.01436.x.

    Article  CAS  PubMed  Google Scholar 

  99. Van Den Buuse M. Circadian rhythms of blood pressure, heart rate, and locomotor activity in spontaneously hypertensive rats as measured with radio-telemetry. Physiol Behav. 1994;55:783–7. https://doi.org/10.1016/0031-9384(94)90060-4.

    Article  PubMed  Google Scholar 

  100. Das UN. Angiotensin-II behaves as an endogenous pro-inflammatory molecule. J Assoc Physicians India. 2005;53:472–6.

    CAS  PubMed  Google Scholar 

  101. Polizio AH, Peña C. Effects of angiotensin II type 1 receptor blockade on the oxidative stress in spontaneously hypertensive rat tissues. Regul Pept. 2005;128:1–5. https://doi.org/10.1016/j.regpep.2004.12.004.

    Article  CAS  PubMed  Google Scholar 

  102. Ambrosio G, Tritto I, Chiariello M. The role of oxygen free radicals in preconditioning. J Mol Cell Cardiol. 1995;27:1035–9. https://doi.org/10.1016/0022-2828(95)90072-1.

    Article  CAS  PubMed  Google Scholar 

  103. Gupta YK, Briyal S. Protective effect of vineatrol against kainic acid induced seizures, oxidative stress and on the expression of heat shock proteins in rats. Eur Neuropsychopharmacol. 2006;16:85–91. https://doi.org/10.1016/j.euroneuro.2005.07.004.

    Article  CAS  PubMed  Google Scholar 

  104. Haugen EN, Croatt AJ, Nath KA. Angiotensin II induces renal oxidant stress in vivo and heme oxygenase-1 in vivo and in vitro. Kidney Int. 2000;58:144–52. https://doi.org/10.1046/j.1523-1755.2000.00150.x.

    Article  CAS  PubMed  Google Scholar 

  105. Zhang H, Schmeißer A, Garlichs CD, Plötze K, Damme U, Mügge A, et al. Angiotensin II-induced superoxide anion generation in human vascular endothelial cells. Role of membrane-bound NADH-/NADPH-oxidases. Cardiovasc Res. 1999;44:215–22. https://doi.org/10.1016/S0008-6363(99)00183-2.

    Article  CAS  PubMed  Google Scholar 

  106. Kazama K, Anrather J, Zhou P, Girouard H, Frys K, Milner TA, et al. Angiotensin II impairs neurovascular coupling in neocortex through NADPH oxidase–derived radicals. Circ Res. 2004;95:1019–26. https://doi.org/10.1161/01.RES.0000148637.85595.c5.

    Article  CAS  PubMed  Google Scholar 

  107. Zimmerman MC, Lazartigues E, Sharma RV, Davisson RL. Hypertension caused by angiotensin II infusion involves increased superoxide production in the central nervous system. Circ Res. 2004;95:210–6. https://doi.org/10.1161/01.RES.0000135483.12297.e4.

    Article  CAS  PubMed  Google Scholar 

  108. De Cavanagh EMV, Piotrkowski B, Fraga CG. Concerted action of the renin-angiotensin system, mitochondria, and antioxidant defenses in aging. Mol Asp Med. 2004;25:27–36. https://doi.org/10.1016/j.mam.2004.02.006.

    Article  CAS  Google Scholar 

  109. Stoynev AG, Ikonomov OC, Minkova NK, Zacharieva SZ, Stoyanovsky VG. Circadian rhythms of arterial pressure: basic regulatory mechanisms and clinical value. Acta Physiol Pharmacol Bulg. 1999;24:43–51.

    CAS  PubMed  Google Scholar 

  110. Campos LA, Bader M, Baltatu OC. Brain renin-angiotensin system in hypertension, cardiac hypertrophy, and heart failure. Front Physiol. 2012;2:115. https://doi.org/10.3389/fphys.2011.00115.

    Article  PubMed  PubMed Central  Google Scholar 

  111. Beĭn G, Shakhotin VN, Muromtseva VM. Diurnal rhythm of the cerebral and systemic circulation in epileptic patients [in Russian]. Zh Nevropatol Psikhiatr Im S S Korsakova. 1978;78:561.

    PubMed  Google Scholar 

  112. Greenwood RS, Meeker R, Sullivan H, Hayward JN. Kindling in spontaneous hypertensive rats. Brain Res. 1989;495:58–65. https://doi.org/10.1016/0006-8993(89)91217-1.

    Article  CAS  PubMed  Google Scholar 

  113. Hernandez CM, Høifødt H, Terry AV. Spontaneously hypertensive rats: further evaluation of age-related memory performance and cholinergic marker expression. J Psychiatry Neurosci. 2003;28:197–209.

    PubMed  PubMed Central  Google Scholar 

  114. Scorza FA, Arida RM, Cysneiros RM, Scorza CA, de Albuquerque M, Cavalheiro EA. Qualitative study of hippocampal formation in hypertensive rats with epilepsy [in Portuguese]. Arq Neuropsiquiatr. 2005;63:283–8.

    Article  Google Scholar 

  115. Meurs A, Clinckersb R, Ebinger G, Michotte Y, Smolders I. Seizure activity and changes in hippocampal extracellular glutamate, GABA, dopamine and serotonin. Epilepsy Res. 2008;78(1):50–9. https://doi.org/10.1016/j.eplepsyres.2007.10.007.

    Article  CAS  PubMed  Google Scholar 

  116. Mendelsohn FA, Jenkins TA, Berkovic SF. Effects of angiotensin II on dopamine and serotonin turnover in the striatum of conscious rats. Brain Res. 1993;613(2):221–9. https://doi.org/10.1016/0006-8993(93)90902-y.

    Article  CAS  PubMed  Google Scholar 

  117. Jobe PC. Common pathogenic mechanisms between depression and epilepsy: an experimental perspective. Epilepsy Behav. 2003;4(Suppl 3):S14–24.

    Article  Google Scholar 

  118. Litt B, Esteller R, Echauz J, D’Alessandro M, Shor R, Henry T, et al. Epileptic seizures may begin hours in advance of clinical onset: a report of five patients. Neuron. 2001;30:51–64. https://doi.org/10.1016/S0896-6273(01)00262-8.

    Article  CAS  PubMed  Google Scholar 

  119. McKeown MJ, McNamara JO. When do epileptic seizures really begin? Neuron. 2001;30:1–3. https://doi.org/10.1016/S0896-6273(01)00253-7.

    Article  CAS  PubMed  Google Scholar 

  120. Arroyo S, de la Morena A. Life-threatening adverse events of antiepileptic drugs. Epilepsy Res. 2001;47:155–74. https://doi.org/10.1016/S0920-1211(01)00306-0.

    Article  CAS  PubMed  Google Scholar 

  121. Berg AT, Langfitt J, Shinnar S, Vickrey BG, Sperling MR, Walczak T, et al. How long does it take for partial epilepsy to become intractable? Neurology. 2003;60:186–90. https://doi.org/10.1212/01.WNL.0000031792.89992.EC.

    Article  CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Natasha Ivanova.

Ethics declarations

Funding

No financial support was received for the publication of this review.

Conflict of interest

Natasha Ivanova and Jana Tchekalarova declare that they have no conflicts of interest.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ivanova, N., Tchekalarova, J. The Potential Therapeutic Capacity of Inhibiting the Brain Renin–Angiotensin System in the Treatment of Co-Morbid Conditions in Epilepsy. CNS Drugs 33, 1101–1112 (2019). https://doi.org/10.1007/s40263-019-00678-4

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s40263-019-00678-4

Navigation