Митохондрии как важная мишень при поиске новых препаратов для лечения болезни Альцгеймера и старческих деменций

##plugins.themes.bootstrap3.article.main##

E.Ф. Шевцова
Д.В. Виноградова
М.Е. Неганова
П.Н. Шевцов
Б.В. Леднев
С.О. Бачурин

Аннотация

Обобщены собственные и литературные данные, обосновывающие роль митохондрий как важнейшей мишени при поиске препаратов для лечения нейродегенеративных заболеваний. Старение является основным фактором риска спорадических форм различных нейродегенеративных заболеваний, в том числе и болезни Альцгеймера (БА). Одной из наиболее аргументированных и принятых в настоящее время является свободнорадикальная митохондриальная теория старения. Именно с ней тесно связаны и митохондриальные гипотезы развития спорадических форм нейродегенеративных заболеваний и, в частности, БА. Нарушение митохондриальных функций приводит к снижению их способности регулировать гомеостаз кальция в клетке и снижению порога для индукции поры митохондриальной проницаемости (МРТ). Ингибиторы МРТ можно рассматривать как перспективный подход к терапии нейродегенеративных заболеваний, так как эти препараты могут не только проявлять свойства нейропротекторов, но и обеспечивать нормализацию синаптической активности благодаря увеличенной кальциевой ёмкости митохондрий. В обзоре представлены данные о ряде ингибиторов МРТ, включая эндогенные соединения – мелатонин, N-ацетилсеротонин, их биоизостерный аналог димебон и ряд других соединений. Использование митохондрий как основы для формирования скрининговой стратегии поиска соединений для лечения нейродегенеративных заболеваний представляет особый интерес и как тестирование их потенциальной токсичности, и как основа для создания метаболических стимуляторов и препаратов, обладающих нейропротекторным и когнитивно-стимулирующим действием.

##plugins.themes.bootstrap3.article.details##

Как цитировать
Шевцова E., Виноградова D., Неганова M., Шевцов P., Леднев B., & Бачурин S. (2018). Митохондрии как важная мишень при поиске новых препаратов для лечения болезни Альцгеймера и старческих деменций. Biomedical Chemistry: Research and Methods, 1(3), e00058. https://doi.org/10.18097/BMCRM00058
Раздел
ОБЗОРЫ

Библиографические ссылки

  1. Qiu C., Kivipelto M., von Strauss E. (2009). Epidemiology of Alzheimer’s disease: occurrence, determinants, and strategies toward intervention. Dialogues Clin. Neurosci, 11(2), 111–128. DOI
  2. Eckert A., Schmitt K., Götz J. (2011). Mitochondrial dysfunction - the beginning of the end in Alzheimer’s disease? Separate and synergistic modes of tau and amyloid-β toxicity. Alzheimers. Res. Ther., 3(2), 15. DOI
  3. Swerdlow R.H., Burns J.M., Khan S.M. (2014). The Alzheimer’s Disease Mitochondrial Cascade Hypothesis: Progress and Perspectives. Biochimica et biophysica acta, 1842(8), 1219-1231. DOI
  4. Caspersen C., Wang N., Yao J., Sosunov A., Chen X., Lustbader J.W., Xu H.W., Stern D., McKhann G., Yan S. Du. (2005). Mitochondrial Abeta: a potential focal point for neuronal metabolic dysfunction in Alzheimer’s disease. FASEB J., 19(14), 2040–2041. DOI
  5. Shevtzova E.F., Kireeva E.G., Bachurin S.O. (2001). Effect of beta-amyloid peptide fragment 25-35 on nonselective permeability of mitochondria. Bull Exp Biol Med., 132(6), 1173-1176. DOI
  6. Kumar A., Bodhinathan K., Foster T.C. (2009). Susceptibility to Calcium Dysregulation during Brain Aging. Front. Aging Neurosci., 1, 2. DOI
  7. Oh M.M., Oliveira F. a., Waters J., Disterhoft J.F. (2013). Altered Calcium Metabolism in Aging CA1 Hippocampal Pyramidal Neurons. J. Neurosci., 33(18), 7905–7911. DOI
  8. Mattson M.P. (2007). Calcium and neurodegeneration. Aging Cell., 6(3), 337–350. DOI
  9. Lopez J.R., Lyckman A., Oddo S., Laferla F.M., Querfurth H.W., Shtifman A. (2008). Increased intraneuronal resting [Ca2+] in adult Alzheimer’s disease mice. J. Neurochem., 105(1), 262–271. DOI 10.1111/j.1471-4159.2007.05135.x
  10. Crompton M. (1999). The mitochondrial permeability transition pore and its role in cell death. Biochem. J., 341, 233–249. DOI
  11. Peng T., Jou M. (2010). Oxidative stress caused by mitochondrial calcium overload., 1201,183–188. DOI 10.1111/j.1749-6632.2010.05634.x
  12. Rapizzi E., Pinton P., Szabadkai G., Wieckowski M.R., Vandecasteele G., Baird G., Tuft R.A., Fogarty K.E., Rizzuto R. (2002). Recombinant expression of the voltage-dependent anion channel enhances the transfer of Ca2+ microdomains to mitochondria. J. Cell Biol., 159(4), 613–624. DOI
  13. Giacomello M., Drago I., Pizzo P., Pozzan T. (2007). Mitochondrial Ca2+ as a key regulator of cell life and death. Cell Death Differ., 14(7), 1267–1274. DOI
  14. Velikanov, G.A. (2013). Endoplasmic reticulum: Membrane contact sites Cell Tiss. Biol., 7, 504-511. https://doi.org/10.1134/S1990519X13060138
  15. Gingrich JR, Pelkey KA, Fam SR, Huang Y, Petralia RS, Wenthold RJ, Salter MW. (2004). Unique domain anchoring of Src to synaptic NMDA receptors via the mitochondrial protein NADH dehydrogenase subunit 2. Proc Natl Acad Sci., 101, 6237–6241. DOI
  16. Shevtsova, E.P., Dubova, L.G., Kireeva, E.G., Bachurin, S.O. (2006). Mitochondria as the memantine target. European Neuropsychopharmacology, 16, 243-244. DOI
  17. Korde A.S., Maragos W.F. Identification of an N-methyl-D-aspartate receptor in isolated nervous system mitochondria. (2012). J Biol Chem., 287(42), 35192-35200. DOI
  18. Bernardi P. (2013). The mitochondrial permeability transition pore: a mystery solved? Front. Physiol. Frontiers, 4, 95. DOI
  19. Weeber E.J., Levy M., Sampson M.J., Anflous K., Armstrong D.L., Brown S.E., Sweatt J.D., Craigen W.J. (2002). The role of mitochondrial porins and the permeability transition pore in learning and synaptic plasticity. J Biol Chem., 277(21), 18891-18897. DOI
  20. Skulachev V.P. (2012). Mitochondria-Targeted Antioxidants as Promising Drugs for Treatment of Age-Related Brain Diseases. J. Alzheimers Dis., 28 (2), 283-289. DOI
  21. Pavlov E., Zakharian E., Bladen C., Diao C.T.M., Grimbly C., Reusch R.N., French R.J. (2005). A large, voltage-dependent channel, isolated from mitochondria by water-free chloroform extraction. Biophys. J., 88(4), 2614–2625. DOI 10.1529/biophysj.104.057281
  22. Abramov A.Y., Fraley C., Diao C.T., Winkfein R., Colicos M.A., Duchen M.R., French R.J., Pavlov E. (2007). Targeted polyphosphatase expression alters mitochondrial metabolism and inhibits calcium-dependent cell death. Proc. Natl. Acad. Sci., 104(46), 18091–18096. DOI
  23. Kokoszka J.E., Waymire K.G., Levy S.E., Sligh J.E., Cai J., Jones D.P., MacGregor G.R., Wallace D.C. (2004). The ADP/ATP translocator is not essential for the mitochondrial permeability transition pore. Nature, 427(6973), 461–465. DOI
  24. Varanyuwatana P., Halestrap A.P. (2012). The roles of phosphate and the phosphate carrier in the mitochondrial permeability transition pore. Mitochondrion, 12, 120–125. DOI
  25. Papadopoulos V., Baraldi M., Guilarte T.R., Knudsen T.B., Lacapère J.-J., Lindemann P., Norenberg M.D., Nutt D., Weizman A., Zhang M.-R., Gavish M. (2006). Translocator protein (18 kDa): new nomenclature for the peripheral-type benzodiazepine receptor based on its structure and molecular function. Trends Pharmacol. Sci., 27(8), 402–409. DOI
  26. Roestenberg P., Manjeri G.R., Valsecchi F., Smeitink J. a M., Willems P.H.G.M., Koopman W.J.H. (2012). Pharmacological targeting of mitochondrial complex I deficiency: the cellular level and beyond. // Mitochondrion, 12(1), 57–65. DOI
  27. Giorgio V., von Stockum S., Antoniel M., Fabbro A., Fogolari F., Forte M., Glick G.D., Petronilli V., Zoratti M., Szabó I., Lippe G., Bernardi P. (2013). ATP synthase dimers form the mitochondrial PTP. PNAS, 110(15), 5887-5892. DOI
  28. Bonora M., Bononi A., De Marchi E., Giorgi C., Lebiedzinska M., Marchi S., Patergnani S., Rimessi A., Suski J.M., Wojtala A., Wieckowski M.R., Kroemer G., Galluzzi L., Pinton P. (2013). Role of the c subunit of the FO ATP synthase in mitochondrial permeability transition. Cell Cycle, 12(4), 674–683. DOI
  29. He J., Ford H.C., Carroll J., Ding S., Fearnley I.M., Walker J.E. (2017). Persistence of the mitochondrial permeability transition in the. absence of subunit c of human ATP synthase. Proc Natl Acad Sci U S A, 114, 3409-3414. DOI
  30. Kowaltowski A.J., Castilho R.F., Vercesi A.E. (2001). Mitochondrial permeability transition and oxidative stress. FEBS Lett., 495(1-2), 12–15. DOI
  31. Luciano M., Patrizia S., Annarita A. (2011). Cyclosporine A in Ullrich Congenital Muscular Dystrophy: Long-Term Results. Oxidative Medicine and Cellular Longevity, 2011, 1-10. DOI
  32. Nighoghossian N., Berthezène Y., Mechtouff L., Derex L., Cho TH., Ritzenthaler T., Rheims S., Chauveau F., Béjot Y., Jacquin A., Giroud M., Ricolfi F., Philippeau F., Lamy C., Turc G., Bodiguel E., Domigo V., Guiraud V., Mas J.L., Oppenheim C., Amarenco P., Cakmak S., Sevin-Allouet M., Guillon B., Desal H., Hosseini H., Sibon I., Mahagne M.H., Ong E., Mewton N., Ovize M. (2015). Cyclosporine in acute ischemic stroke. Neurology., 84(22), 2216-23. DOI
  33. Atkinson K., Britton K., Biggs J. (1984). Distribution and concentration of cyclosporin in human blood. Journal of Clinical Pathology., 37(10), 1167-1171. DOI
  34. Readnower R.D., Pandya J.D., McEwen M.L., Pauly J.R., Springer J.E., Sullivan P.G. (2011). Post-injury administration of the mitochondrial permeability transition pore inhibitor, NIM811, is neuroprotective and improves cognition after traumatic brain injury in rats. J. Neurotrauma, 28(9), 1845–1853. DOI
  35. Korde A. S., Pettigrew L. C., Craddock S. D., Pocernich C. B., Waldmeier P. C., Maragos W. F. (2007). Protective Effects of NIM811 in Transient Focal Cerebral Ischemia Suggest Involvement of the Mitochondrial Permeability Transition. J. Neurotrauma, 24, 895−908. DOI
  36. Wissing E. R., Millay D. P., Vuagniaux G., Molkentin J. D. (2010). Debio-025 Is More Effective Than Prednisone in Reducing Muscular Pathology in Mdx Mice. Neuromuscul. Disord., 20, 753−760. DOI
  37. Warne J., Pryce G., Hill J.M., Shi X., Lennerås F., Puentes F., Kip M., Hilditch L., Walker P., Simone M.I., Chan A.W., Towers G.J., Coker A.R., Duchen M.R., Szabadkai G., Baker D., Selwood D.L. (2016). Selective Inhibition of the Mitochondrial Permeability Transition Pore Protects against Neurodegeneration in Experimental Multiple Sclerosis. J Biol Chem., 291(9), 4356-4373. DOI
  38. Roy S., Šileikytė J., Schiavone M., Neuenswander B., Argenton F., Aubé J., Hedrick M.P., Chung T.D., Forte M.A., Bernardi P., Schoenen F.J. (2015). Discovery, Synthesis, and Optimization of Diarylisoxazole-3-carboxamides as Potent Inhibitors of the Mitochondrial Permeability Transition Pore. ChemMedChem., 10(10), 1655-71. DOI
  39. Averina E.B., Gracheva Y.A., Grishin Y.K., Radchenko E.V., Burmistrov V.V., Butov G.M., Neganova M.E., Serkova T.P., Redkozubova O.M., Shevtsova E.F., Milaeva E.R., Kuznetsova T.S., Zefirov N.S. (2016). Synthesis and biological evaluation of novel 5-hydroxylaminoisoxazole derivatives as lipoxygenase inhibitors and metabolism enhancing agents. Bioorganic & Medicinal Chemistry, 24(4), 712-720. DOI
  40. Smith R.A., Adlam V.J., Blaikie F.H., Manas A.R., Porteous C.M., James A.M., Ross M.F., Logan A., Cochemé H.M., Trnka J., Prime T.A., Abakumova I., Jones B.A., Filipovska A., Murphy M.P. (2008). Mitochondria-targeted antioxidants in the treatment of disease. Ann N Y Acad Sci., 1147, 105-111. DOI
  41. Rocha M., Hernandez-Mijares A., Garcia-Malpartida K., Bañuls C., Bellod L., Victor V.M. (2010). Mitochondria-targeted antioxidant peptides. Curr Pharm Des., 16(28), 3124-3131. DOI
  42. Srinivasan V., Spence D.W., Pandi-Perumal S.R., Brown G.M., Cardinali D.P. (2011). Melatonin in Mitochondrial Dysfunction and Related Disorders. International Journal of Alzheimer’s Disease, 2011, 1-16. DOI
  43. He H., Dong W., Huang F. (2010). Anti-amyloidogenic and anti-apoptotic role of melatonin in Alzheimer disease. Curr Neuropharmacol., 8(3), 211-217. DOI
  44. Pandi-Perumal S.R., BaHammam A.S., Brown G.M., Spence D.W., Bharti V.K., Kaur C., Hardeland R., Cardinali D.P. (2013). Melatonin antioxidative defense: therapeutical implications for aging and neurodegenerative processes. Neurotox Res., 23(3), 267-300. DOI
  45. Andrabi S.A., Sayeed I., Siemen D., Wolf G., Horn T.F. (2004). Direct inhibition of the mitochondrial permeability transition pore: a possible mechanism responsible for anti-apoptotic effects of melatonin. FASEB J., 18(7), 869-871. DOI
  46. Bachurin S., Oxenkrug G., Lermontova N., Afanasiev A., Beznosko B., Vankin G., Shevtsova E., Mukhina T., Serkova T. (2000). N-Acetyl-Serotonin, Melatonin and Their Derivatives Improve Cognition and Protect Against beta-Amyloid-induced Neurotoxicity. Annals of NY Aca of Sci., 890, 156-166. DOI
  47. Zhou H., Wang J., Jiang J., Stavrovskaya I.G., Li M., Li W., Wu Q., Zhang X., Luo C., Zhou S., Sirianni A.C., Sarkar S., Kristal B.S., Friedlander R.M., Wang X. (2014). N-acetyl-serotonin offers neuroprotection through inhibiting mitochondrial death pathways and autophagic activation in experimental models of ischemic injury. J Neurosci., 34(8), 2967-2978. DOI
  48. Wang X., Figueroa B.E., Stavrovskaya I.G., Zhang Y., Sirianni A.C., Zhu S., Day A.L., Kristal B.S., Friedlander R.M. (2009). Methazolamide and melatonin inhibit mitochondrial cytochrome C release and are neuroprotective in experimental models of ischemic injury. Stroke, 40(5), 1877-85. DOI
  49. Bachurin S.O., Shevtsova E.P., Kireeva E.G., Oxenkrug G.F., Sablin S.O. (2003). Mitochondria as a target for neurotoxins and neuroprotective agents. Ann N Y Acad Sci., 993, 334-344. DOI
  50. Millán-Plano Sergio, Eduardo Piedrafita, Francisco J. Miana-Mena , Lorena Fuentes-Broto. (2010). Melatonin and Structurally-Related Compounds Protect Synaptosomal Membranes from Free Radical Damage. J. Mol. Sci., 11, 312-328. DOI
  51. Neganova M.E., Klochkov S.G., Afanasieva S.V., Chudinova E.S., Serkova T.P., Shevtsova E.F. (2014). Allomargaritarine as a basis for the creation of mitochondrial targeted potential neuroprotectors. Eur Neuropsychopharmacol., 24, 262. DOI
  52. Lin X., Jun-Tian Z. (2004). Neuroprotection by D-securinine against neurotoxicity induced by beta-amyloid (25-35). Neurol Res., 26(7), 792-796. DOI
  53. Raj D., Luczkiewicz M. (2008). Securinega suffruticosa. Fitoterapia, 79(6), 419-427. DOI
  54. Neganova M.E., Serkova T.P., Klochkov S.G., Afanasieva S.V., Shevtsova E.F., Bachurin S.O. (2011). Neuroprotective properties of Allomargaritarine, a Novel Tryptamine Derivative of the Natural Alkaloid Securinine Natural and Technical Sciences., 5, 86-90.
  55. Neganova M.E., Klochkov S.G., Afanasieva S.V., Serkova T.P., Chudinova E.S., Bachurin S.O., Reddy V.P., Aliev G., Shevtsova E.F. (2016). Neuroprotective effects of the securinine-analogues: identification of Allomargaritarine as a lead compound CNS & Neurological Disorders - Drug Targets, 15, 102-107. DOI : 10.2174/1871527314666150821111812
  56. Neganova M.E., Klochkov S.G., Petrova L.N., Shevtsova E.F., Afanasieva S.V., Chudinova E..S, Fisenko V.P., Bachurin S.O., Barreto G.E., Aliev G. (2017). Securinine Derivatives as Potential Anti-amyloid Therapeutic Approach. CNS Neurol Disord Drug Targets, 16(3), 351-355. DOI
  57. Neganova M.E., Blik V.A., Klochkov S.G., Chepurnova N.E., Shevtsova E.F. (2011). Investigation of the Antioxidant Characteristics of a New Tryptamine Derivative of Securinine and its Influence on Seizure Activity in the Brain in Experimental Epilepsy. Neurochemical Journal, 5, 208. DOI
  58. Kim do Y., Simeone K.A., Simeone T.A., Pandya J.D., Wilke J.C., Ahn Y., Geddes J.W., Sullivan P.G., Rho J.M. (2015). Ketone bodies mediate antiseizure effects through mitochondrial permeability transition. Ann Neurol., 78(1), 77-87. DOI
  59. Kudin A.P., Debska-Vielhaber G., Vielhaber S., Elger C.E., Kunz W.S. (2004). The mechanism of neuroprotection by topiramate in an animal model of epilepsy. Epilepsia, 45(12), 1478-1487. DOI
  60. Matveeva I.A. (1983). Action of dimebon on histamine receptors. Pharmakology and Toxicology, 46(4), 27–29. DOI
  61. Bachurin S.O., Shevtsova E.P., Kireeva E.G., Oxenkrug G.F., Sablin S.O. (2003). Mitochondria as a Target for Neurotoxins and Neuroprotective Agents. Ann. N.Y. Acad. Sci., 993, 334–344. DOI
  62. Shevtsova E.F., Vinogradova D.V., Kireeva E.G., V. Prakash Reddy, Aliev G., Bachurin S.O. (2015). Dimebon Attenuates the Rat-Brain Mitochondrial Permeabilization. Current Alzheimer Research, 11(5), 422-429. DOI
  63. Zhang S., Hedskog L., Petersen C.A., Winblad B., Ankarcrona M. (2010). Dimebon (latrepirdine) enhances mitochondrial function and protects neuronal cells from death. J Alzheimers Dis., 21(2), 389-402. DOI
  64. Bharadwaj P.R, Verdile G., Barr R.K., Gupta V., Steele J.W., Lachenmayer M.L., Yue Z., Ehrlich M.E., Petsko G., Ju S., Ringe D., Sankovich S.E., Caine J.M., Macreadie I.G., Gandy S., Martins R.N. (2012). Latrepirdine (dimebon) enhances autophagy and reduces intracellular GFP-Aβ42 levels in yeast. J Alzheimers Dis., 32(4), 949-67. DOI
  65. Eckert S.H., Eckmann J., Renner K., Eckert G..P, Leuner K., Muller W.E. (2012). Dimebon ameliorates amyloid-β induced impairments of mitochondrial form and function. J Alzheimers Dis., 31(1), 21-32. DOI
  66. Eckert S., Gaca J., Kolesova N. (2018). Mitochondrial Pharmacology of Dimebon (Latrepirdine) Calls for a New Look at its Possible Therapeutic Potential in Alzheimer’s Disease J. Aging Dis., 9(4), 729-744. DOI
  67. Bachurin, V. Grigoriev, E. Shevtsova, I. Koroleva, L. Dubova, E. Kireeva. (2007). Anti-aging properties of Dimebon: Relation to mitochondrial permeability inhibition, Experimental Gerontology, 42(1–2), 142-143. DOI
  68. Peters O.M., Shelkovnikova T., Tarasova T., Springe S., Kukharsky M.S., Smith G.A., Brooks S., Kozin S.A., Kotelevtsev Y., Bachurin S.O., Ninkina N., Buchman V.L. (2013). Chronic administration of Dimebon does not ameliorate amyloid-β pathology in 5xFAD transgenic mice. J Alzheimers Dis., 36(3), 589-596. DOI
  69. Peters O.M, Connor-Robson N., Sokolov V.B., Aksinenko A.Y., Kukharsky M.S., Bachurin S.O., Ninkina N., Buchman V.L. (2013). Chronic administration of dimebon ameliorates pathology in TauP301S transgenic mice. J Alzheimers Dis., 33(4), 1041-1049. DOI
  70. Bachurin S.O., Shelkovnikova T.A., Ustyugov A.A., Peters O., Khritankova I., Afanasieva M.A., Tarasova T.V., Alentov I.I., Buchman V.L., Ninkina N.N. (2012). Dimebon slows progression of proteinopathy in γ-synuclein transgenic mice. Neurotox Res., 22(1), 33-42. DOI
  71. Ustyugov A.A., Shelkovnikova T.A., Kokhan V.S., Khritankova I..V, Peters O., Buchman V.L., Bachurin S.O., Ninkina N.N. (2012). Dimebon reduces the levels of aggregated amyloidogenic protein forms in detergent-insoluble fractions in vivo. Bull Exp Biol Med., 152(6), 731-733. DOI
  72. Yamashita M., Nonaka T., Arai T., Kametani F., Buchman V.L., Ninkina N., Bachurin S.O., Akiyama H., Goedert M., Hasegawa M. (2009). Methylene blue and dimebon inhibit aggregation of TDP-43 in cellular models. FEBS Lett., 583(14), 2419-2424. DOI
  73. Khritankova I.V., Kukharskiy M.S., Lytkina O.A., Bachurin S.O., Shorning B.Y. (2012). Dimebon activates autophagosome components in human neuroblastoma SH-SY5Y cells. Dokl Biochem Biophys., 446, 251-253. DOI
  74. Pieper A.A, Xie S., Capota E., Estill S.J., Zhong J., Long J.M., Becker G.L., Huntington P., Goldman S.E., Shen C.H., Capota M., Britt J.K., Kotti T., Ure K., Brat D.J., Williams N.S., MacMillan K.S., Naidoo J., Melito L., Hsieh J., De Brabander J., Ready J.M., McKnight S.L. (2010). Discovery of a proneurogenic, neuroprotective chemical. Cell, 142(1), 39-51. DOI
  75. Hou Y., Mattson M.P., Cheng A. (2013). Permeability Transition Pore-Mediated Mitochondrial Superoxide Flashes Regulate Cortical Neural Progenitor Differentiation. PLoS ONE, 8(10), 76721. DOI
  76. Shin J.Y., Kong S.Y., Yoon H.J., Ann J., Lee J., Kim H.J. (2015). An Aminopropyl Carbazole Derivative Induces Neurogenesis by Increasing Final Cell Division in Neural Stem Cells. Biomol Ther (Seoul), 23(4), 313-319. DOI
  77. Wang G., Han T., Nijhawan D., Theodoropoulos P., Naidoo J., Yadavalli S., Mirzaei H., Pieper A.A., Ready J.M., McKnight S.L. (2014). P7C3 neuroprotective chemicals function by activating the rate-limiting enzyme in NAD salvage. Cell, 158(6), 1324-1334. DOI
  78. Wang S.N., Xu T.Y., Wang X., Guan Y.F., Zhang S.L., Wang P., Miao C.Y. (2016). Neuroprotective Efficacy of an Aminopropyl Carbazole Derivative P7C3-A20 in Ischemic Stroke. CNS Neurosci Ther, 22(9), 782-788. DOI
  79. Jiang B., Song L., Huang C., Zhang W. (2016). P7C3 Attenuates the Scopolamine-Induced Memory Impairments in C57BL/6J Mice. Neurochem Res., 41(5), 1010-1019. DOI
  80. Blaya M.O., Bramlett H.M., Naidoo J., Pieper A.A., Dietrich W.D. (2014). Neuroprotective efficacy of a proneurogenic compound after traumatic brain injury. J Neurotrauma, 31(5), 476-486. DOI
  81. Kuharskiy M.S., Ovchinnikov R.K., Ustyugov A.A., Bachurin S.O. (2014). Molecular Aspects of Pathogenesis and Modern Appoaches to Pharmacological Correction of Alzheimer's Disease. Neurodegenerative Diseases: from the Genome to the Whole Organism, Ed. M.V.Ugryumov, Moscow, Scientific World, 2, 137-162.
  82. Bachurin S. (2015). Contemporary Approaches for Pharmacological Intervention of Abundant Neurodegenerative Disorders. J Nanomedine Biotherapeutic Discov, 5(2), 137. DOI