ИНТРАНАЗАЛЬНОЕ ВВЕДЕНИЕ ИНСУЛИНА КРЫСАМ С ИШЕМИЕЙ И РЕПЕРФУЗИЕЙ ПЕРЕДНЕГО МОЗГА УМЕНЬШАЕТ ИНТЕНСИВНОСТЬ АУТОФАГИИ И АПОПТОЗА В ГИППОКАМПЕ И ЛОБНОЙ КОРЕ МОЗГА, ВОЗМОЖНЫЙ МЕХАНИЗМ ДЕЙСТВИЯ
PDF

Ключевые слова

ишемия и реперфузия переднего мозга
аутофагия
апоптоз
интраназальное введение инсулина
протеинкиназы

Аннотация

Найдено, что ишемия и последующая трехдневная реперфузия переднего мозга приводят к увеличению уровня маркера аутофагии LC3B-II, уровня глиального фибриллярного кислого белка (GFAP) и активации каспазы-3 в гиппокампе и лобной коре мозга крыс. При этом интраназальное введение крысам с ишемией и реперфузией мозга 0.5 МЕ инсулина (до ишемии и ежедневно при реперфузии) достоверно и значительно снижает уровень LC3B-II и активность каспазы-3 в изучаемых структурах. Это показывает способность инсулина ингибировать активацию аутофагии и апоптоза в переднем мозге при его ишемии и реперфузии. Не удалось выявить достоверного снижения уровня GFAP в этих районах мозга под влиянием введения животным инсулина. Показано, что интраназальное введение инсулина активирует протеинкиназу Akt (активирующую комплекс mTORС1, ингибирующий процессы аутофагии) и ингибирует протеинкиназу AMPK (инициируюшую процессы аутофагии) в гиппокампе и коре мозга крыс, что, очевидно, лежит в основе его способности снижать аутофагическую и апоптотическую гибель нейронов. Данные о модуляции инсулином активности протеинкиназ Akt и AMPK в опытах in vivo согласуются с результатами исследованиями возможного механизма нейропротекторного действия инсулина, проведенными нами ранее in vitro на нейронах коры мозга в состоянии окислительного стресса.

https://doi.org/10.31857/S0044452924010053
PDF

Литература

GBD 2019 Stroke Collaborator (2021). Global, regional, and national burden of stroke and its risk factors, 1990-2019: a systematic analysis for the Global Burden of Disease Study 2019. Lancet Neurol 20 (10):795–820. https://doi.org/10.1016/S1474-4422(21)00252-0

Kuriakose D, Xiao Z (2020) Pathophysiology and treatment of stroke: Present status and future perspectives. Int J Mol Sci 21 (20): 7609. https://doi.org/10.3390/ijms21207609

Barthels D, Das H (2020) Current advances in ischemic stroke research and therapies. Biochim Biophys Acta. Mol Basis Dis 1866 (4): 165260. http://doi.org/10.1016/j.bbadis.2018.09.012

Cannistraro RJ, Badi M, Eidelman BH, Dickson DW, Middlebrooks EH, Meschia JF (2019) CNS small vessel disease: A clinical review. Neurology: 92 (24) 1146–1156. http://doi.org/10.1212/WNL.0000000000007654

Han F, Zhang DD, Zhai FF, Xue J, Zhang JT, Yan S, Zhou LX, Ni J, Yao M, Yang M, Li ML, Jin ZY, Dai Q, Zhang SY, Cui LY, Zhu YC (2021) Association between large artery stenosis, cerebral small vessel disease and risk of ischemic stroke. .Sci China Life Sci: 64 (9) 1473–1480. http://doi.org/10.1007/s11427-020-849-x

Markus HS, de Leeuw FE (2023) Cerebral small vessel disease: Recent advances and future directions. Int J Stroke: 18 (1) 4–14. http://doi.org/10.1177/17474930221144911

Fan LW, Carter K, Beatt A, Pang Y (2019) Rapid transport of insulin to the brain following intranasal administration in rats. Neural Regen Res 14: 1046–1051. https://doi.org/10.4103/1673-5374.250624

Tashima T (2020) Shortcut approaches to substance delivery into the brain based on intranasal administration using nanodelivery strategies for insulin. Molecules 25 (21): 5188. https://doi.org/10.3390/molecules25215188

Craft S, Claxton A, Baker LD, Hanson AJ, Collerton B, Trittschuh EH, Dahl D, Caulder E, Neth B, Montine TJ, Jung Y, Maldjian J, Whitlow C, Friedman S (2017) Effects of regular and long-acting insulin on cognition and Alzheimer's disease biomarkers: A pilot clinical trial. J Alzheimers Dis 57: 1325–1334. https://doi.org/10.3233/JAD-1612566

Avgerinos KI, Kalaitzidis G, Malli A, Kalaitzoglou D, Myserlis PG, Lioutas VA (2018) Intranasal insulin in Alzheimer’s dementia or mild cognitive impairment. A systematic review. J Neurol 265: 1497–1510. https://doi.org/10.1007/s00415-018-8768-0

Novak P, Maldonado DAP, Novak V (2019) Safety and preliminary efficacy of intranasal insulin for cognitive impairment in Parkinson disease and multiple system atropA double-blinded placebo-controlled pilot study. PLoS One 14: e0214364. https://doi.org/10.1371/journal.pone.0214364

Guo Z, Chen Y, Mao YF, Zheng T, Jiang Y, Yan Y, Yin X, Zhang B (2017) Long-term treatment with intranasal insulin ameliorates cognitive impairment, tau hyperphosphorylation, and microglial activation in a streptozotocin-induced Alzheimer’s rat model. Sci Rep 7: 45971. https://doi.org/ 10.1038/srep45971

Kamat PK, Kalani A, Rai S, Tota SK, Kumar A, Ahmad AS (2016) Streptozotocin intracerebroventricular-induced neurotoxicity and brain insulin resistance: a therapeutic intervention for treatment of sporadic Alzheimer's disease (sAD)-like pathology. Mol Neurobiol 53 (7): 4548–4562. https:/ doi.org/10.1007/s12035-015-9384-y

Zorina II, Zakharova IO, Bayunova LV, Avrova NF (2018) Insulin administration prevents accumulation of conjugated dienes and trienes and tnactivation of Na+, K+-ATPase in the rat cerebral cortex during two-vessel forebrain ischemia and reperfusion. J Evol Biochem Phys 54 (3): 246–249. https://doi.org/10.1371/journal.pone.0214364

Zorina II, Fokina EA, Zakharova IO, Bayunova LV, Shpakov AO (2019) Features of the changes in lipid peroxidation and activity of Na+/K+-ATPase in the brain of the aged rats in the conditions of two-vessel cerebral ischemia/reperfusion. Adv Gerontol. 32 (6): 941–947.

Nabavi SF, Sureda A, Sanches-Silva A, Pandima DK, Ahmed T, Shahid M (2019) Novel therapeutic strategies for stroke: the role of autophagy. Critical Reviews in Clinical Laboratory Sciences 56(3): 182–199. https://doi.org/10.1080/10408363.2019.1575333

Campbell BCV, Khatri P (2020) Stroke. Lancet 396 (10244): 149-142. https://doi.org/10.1016/S0140-6736(20)31179-X

He C, Xu Y, Sun J, Li L, Zhang JH, Wang Y (2022) Autphagy and apoptosis in acute brain injuries: From mechanism to treatment. Antioxid Redox Signal. https://doi.org/10.1089/ars.2021.0094

Zhou H, Wang J, Jiang J, Stavrovskaya IG, Li M, Li W, Wu Q, Zhang X, Luo C, Zhou S, Sirianni AC, Sarkar S, Kristal BS, Friedlander RM, 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: 2967–2978. https://doi.org/10.1523/JNEUROSCI.1948-13.2014

Wang M, Li Y.-J, Ding Y, Zhang H-N, Sun T, Zhang K, Yang L, Guo Y-Y, Liu S-B, Zhao M-G, Qu Y-M (2016) Silibinin prevents autophagic cell death upon oxidative stress in cortical neurons and cerebral ischemia-reperfusion injury. Mol Neurobiol 53: 932–943. https://doi.org/10.1007/s12035-014-9062-5

Li L, Tian J, Long MK-W, Chen Y, Lu J, Zhou C, Wang T (2016) Protection against experimental stroke by ganglioside GM1 is associated with the inhibition of autophagy. PLoS One 11: e0144219. https://doi.org/10.1371/journal.pone.0144219

Li X, Wang M, Qin C, Fan W-H, Tian D-S, Liu J-L (2017) Fingolimod suppresses neuronal autophagy through the mTOR/p70S6K pathway and alleviates ischemic brain damage in mice. PloS One 12: e0188748. https://doi.org/10.1371/journal.pone.0188748

Hu Y, Zhou H, Zhang H, Sui Y, Zhang Z, Zou Y, Li K, Zhao Y, Xie J, Zhang L (2022) The neuroprotective effect of dexmedetomidine and its mechanism. Front Pharmacol 13: 965661. eCollection 2022. https://doi.org/10.3389/fphar.2022.965661

Zhang H, Wang X, Chen W, Yang Y, Wang Y, Wan H, Zhu Z (2023) Danhong injection alleviates cerebral ischemia-reperfusion injury by inhibiting autophagy through miRNA-132-3p/ATG12 signal axis. J Ethnopharmacol 300:115724. https://doi.org/10.1016/j.jep.2022.115724

Buckley KM, Hess DL, Sazonova IY, Periyasamy-Thandavan S, John R, Barrett JR, Kirks R, Grace H, Kondrikova G, Johnson MH, Hess DC, Schoenlein PV, Hoda MN, Hill WD (2014) Rapamycin up-regulation of autophagy reduces infarct size and improves outcomes in both permanent MCAL, and embolic MCAO, murine models of stroke. Exp Transl Stroke Med 6: 8. eCollection. https://doi.org/10.1186/2040-7378-6-8

Liu X, Tian F, Wang S, Wang F, Xiong L (2018) Astrocyte Autophagy Flux Protects Neurons Against Oxygen-Glucose Deprivation and Ischemic/Reperfusion Injury. Rejuvenation Res 215: 405–415. https://doi.org/10.1089/rej.2017.1999

Carloni S, Balduini W (2020) Simvastatin preconditioning confers neuroprotection against hypoxia-ischemia induced brain damage in neonatal rats via autophagy and silent information regulator 1 (SIRT1) activation. Exp Neurol 324: 113117. https://doi.org/10.1016/j.expneurol.2019.113117

Fokina EA, Zakharova IO, Bayunova LV, Avrova DK, Ilyasov IO, Avrova NF (2023) Intranasal insulin decreases autophagic and apoptotic death of neurons in the rat hippocampal CA1 region and frontal cortex under forebrain ischemia-reperfusion. J Evol Biochem Physiol 59 (1): 45–56. https://doi.org/10.1134/S0022093023010040

Zakharova IO, Bayunova LV,Avrova DK, Avrova NF (2023) Neuroprotective effect of insulin on rat cortical neurons in oxidative stress is mediated by autophagy and apoptosis inhibition in vitro. J Evol Biochem Phys 59 (5): 1536–1550. https://doi.org/10.1134/S0022093023050071

Paxinos G, Watson C (1998) The rat brain in stereotaxic coordinates. Fourth Edition. Acad. Press. San Diego Calif. USA 1–237.

Молчанова СМ, Москвин АН, Захарова ИО, Юрлова ЛА, Носова ИЮ, Аврова НФ (2005) Влияние двухсосудистой ишемии переднего мозга и введения индометацина и квинакрина на активность Na+,K+-АТФ-азы в разных областях мозга крыс. Ж эвол биохим физиол 41(1): 33–38. [Molchanova SM, Moskvin AN, Zakharova IIu, Iurlova LA, Nosova IIu, Avrova NF (2005) Na,K-atpase activity in different brain regions in cerebral ischemia and influence of quinacrine and indomethacin administration. Zh Evol Biokhim Fiziol 41(1):33–38. (In Russ)].

Sanderson TH, Wider JM (2013) 2-Vessel occlusion/hypotension: A rat model of global brain ischemia. J Vis Exp 76: e50173. https://doi.org/10.3791/50173

Zakharova IO, Sokolova TV, Vlasova YA, Bayunova LV, Rychkova MP, Avrova NF (2017) α-Tocopherol at nanomolar concentration protects cortical neurons against oxidative stress. Int J Mol Sci 18: 216. https://doi.org/10.3390/ijms18010216

Zick Y (2005) Ser/Thr phosphorylation of IRS proteins: a molecular basis for insulin resistance Sci STKE 268:pe4. https://doi.org/10.1126/stke.2682005pe4

Tanida I, Ueno T, Kominami E (2004) LC3 conjugation system in mammalian autophagy. Int J Biochem Cell Biol 36(12):2503-2518. https://doi.org/10.1016/j.biocel.2004.05.009

Fritzen AM, Frøsig C, Jeppesen J, Jensen TE, Lundsgaard AM, Serup AK, Schjerling P, Proud CG, Richter EA, Kiens B (2016) Role of AMPK in regulation of LC3 lipidation as a marker of autophagy in skeletal muscle. Cell Signal 28(6):663–674. https://doi.org/10.1016/j.cellsig.2016.03.005

Bansal M, Moharir SC, Swarup G (2018) Autophagy receptor optineurin promotes autophagosome formation by potentiating LC3-II production and phagophore maturation. Commun Integr Biol 11 (2): 1–4. https://doi.org/10.1080/19420889.2018.1467189

Ribeiro M, López de Figueroa P, Blanco FJ, Mendes AF, Caramés B (2016) Insulin decreases autophagy and leads to cartilage degradation. Osteoarthritis Cartilage 24:731–739. https://doi.org/10.1016/j.joca.2015.10.017

Pires KM, Torres NS, Buffolo M, Gunville R, Schaaf C, Davis K, Selzman CH, Gottlieb RA, Boudina S (2019) Suppression of cardiac autophagy by hyperinsulinemia in insulin receptor-deficient hearts is mediated by insulin-like growth factor receptor signaling. Antioxid Redox Signal 31(6):444–457. https://doi.org/10.1089/ars.2018.7640

Ding X, Zhang L, Zhang X, Qin Y, Yu K, Yang X (2023) Intranasal insulin alleviates traumatic brain injury by inhibiting autophagy and endoplasmic reticulum stress-mediated apoptosis through the PI3K/Akt/mTOR signaling pathway. Neuroscience 29:23–36. https://doi.org.10.1016/j.neuroscience.2023.08.009

Sanderson TH, Kumar R, Murariu-Dobrin AC, Page AB, Krause GS, Sullivan JM (2009) Insulin activates the PI3K-Akt survival pathway in vulnerable neurons following global brain ischemia. Neurol Res. 31(9):947–958. https://doi.org/10.1179/174313209X382449

Russo V, Candeloro P, Malara N, Perozziello G, Iannone M, Scicchitano M, Mollace R, Musolino V, Gliozzi M, Carresi C (2019) Key role of cytochrome C for apoptosis detection using Raman microimaging in an animal model of brain ischemia with insulin treatment. Appl Spectrosc 73 (10): 1208–1217. https://doi.org/10.1177/0003702819858671

Zhu Y-M, Gao X, Ni Y, Li W, Ken TA, Qiao S-G, Wang C, Xiao-Xuan Xu X-X, Hui-Ling Zhang H-L (2017) Sevoflurane postconditioning attenuates reactive astrogliosis and glial scar formation after ischemia-reperfusion brain injury. Neuroscience 356:125–141. https://doi.org/10.1016/j.neuroscience.2017.05.004

Zhang R, Pei H, Ru L, Li H, Liu G (2013) Bone morphogenetic protein 7 upregulates the expression of nestin and glial fibrillary acidic protein in rats with cerebral ischemia-reperfusion injury. Biomed Rep 1(6): 895–900. https://doi.org/10.3892/br.2013.164

Yamada M, Ohnishi H, Sano SA, Nakatani A, Ikeuchi T, Hatanaka H (1997) Insulin receptor substrate IRS-1 and IRS-2 are tyrosine-phosphorylated and associated with phosphatidylinositol 3-kinase in response to brain-derived neurotrophic factor in cultured cerebral cortical neurons. J Biol Chem 272(48):30334–30339. https://doi.org/10.1074/jbc.272.48.30334

Grote CW, Morris JK, Ryals JM, Geiger PC, Wright DE (2011). Insulin receptor substrate 2 expression and involvement in neuronal insulin resistance in diabetic neuropathy. Exp Diabetes Res 2011: 212571. https://doi. org/10.1155/2011/21257

Boura-Halfon S, Zick T (2009) Phosphorylation of IRS proteins, insulin action, and insulin resistance. Am J Physiol Endocrinol Metab 296(4): E581-591. https://doi.org/10.1152/ajpendo.90437.2008

Li Y, Chen Y (2019) AMPK and autophagy. Adv Exp Med Biol 1206: 85–108. https://doi.org/10.1007/978-981-15-0602-4_4

Kim J, Kundu M, Viollet B, Guan K-L (2011) AMPK and mTOR regulate autophagy through direct phosphorylation of Ulk1. Nat Cell Biol 13 (2):132–141. https://doi.org/10.1038/ncb2152

Parzych KR, Klionsky DJ (2014) An overview of autophagy: morphology, mechanism, and regulation. Antioxid Redox Signals 20(3):460–473. https://doi. org/:10.1089/ars.2013.5371

Yoon MS (2017) The role of mammalian target of rapamycin (mTOR) in insulin signaling, nutrients. 9 (11): 1176. https://doi. org/ 10.3390/nu9111176

Sharma A, Anand SK, Singh N, Dwivedi UN, Kakkar P (2023) AMP-activated protein kinase: An energy sensor and survival mechanism in the reinstatement of metabolic homeostasis. Exp Cell Res 428(1):113614. https://doi.org/10.1016/j.yexcr.2023.113614

Yang K, Chen Z, Gao J, Shi W, Li L, Jiang S, Hu H, Liu Z, Xu D, Wu L (2017) The key roles of GSK-3beta in regulating mitochondrial activity. Cell Physiol Biochem 44:1445–1459. https://doi.org/10.1159/000485580