doi: 10.3967/bes2024.112
Nogo-A Protein Mediates Oxidative Stress and Synaptic Damage Induced by High-altitude Hypoxia in the Rat Hippocampus
-
Abstract:
Objective High-altitude hypoxia exposure often damages hippocampus-dependent learning and memory. Nogo-A is an important axonal growth inhibitory factor. However, its function in high-altitude hypoxia and its mechanism of action remain unclear. Methods In an in vivo study, a low-pressure oxygen chamber was used to simulate high-altitude hypoxia, and genetic or pharmacological intervention was used to block the Nogo-A/NgR1 signaling pathway. Contextual fear conditioning and Morris water maze behavioral tests were used to assess learning and memory in rats, and synaptic damage in the hippocampus and changes in oxidative stress levels were observed. In vitro, SH-SY5Y cells were used to assess oxidative stress and mitochondrial function with or without Nogo-A knockdown in Oxygen Glucose-Deprivation/Reperfusion (OGD/R) models. Results Exposure to acute high-altitude hypoxia for 3 or 7 days impaired learning and memory in rats, triggered oxidative stress in the hippocampal tissue, and reduced the dendritic spine density of hippocampal neurons. Blocking the Nogo-A/NgR1 pathway ameliorated oxidative stress, synaptic damage, and the learning and memory impairment induced by high-altitude exposure. Conclusion Our results demonstrate the detrimental role of Nogo-A protein in mediating learning and memory impairment under high-altitude hypoxia and suggest the potential of the Nogo-A/NgR1 signaling pathway as a crucial therapeutic target for alleviating learning and memory dysfunction induced by high-altitude exposure. -
Key words:
- Nogo-A /
- NgR1 /
- High-altitude hypoxia /
- Learning and memory /
- Oxidative stress
注释:1) CONFLICT OF INTEREST: -
Figure 1. The effects of HH exposure on contextual fear conditioning test and Morris Water Maze (MWM) behavior in rats. (A) Schematic illustration of the strategy for a contextual fear conditioning test. (B) Freezing time in context test was measured after electric shock training. (Bars represent mean ± sem. Student’s t-test, *P<0.05, N=10). (C) Schematic illustration of the strategy for the MWM test pattern. (D) Escape distance to the platform, (E) mean swimming speed of the rats in the training phase, (F) primary escape latency, (G) time spent in the target quadrant, and (H) number of target entries of rats in the test phase of the MWM. (I) Representative picture of a rat track plot in the MWM test phase (data are shown as mean ± sem, N=12, Student’s t-test, *P<0.05, **P< 0.01, ***P < 0.001, ns, not significant). (J) Quantification of ROS levels, (K) MDA levels, (L) SOD activity, and (M) ratio of GSH-to-GSSG in hippocampus homogenates after HH exposure. (Bars represent mean ± sem. Statistical significance was assessed using Student’s t-test. *P < 0.05, **P< 0.01, ***P < 0.001, N = 6).
Figure 2. The effects of HH exposure on the expression of Nogo-A and NgR1 proteins in the hippocampal region and synaptic damage in hippocampal neurons. (A) Representative western blot of Nogo-A, NgR1, S1PR2, and β-actin in rat hippocampal lysates. (B) The quantification of Nogo-A, NgR1, and S1PR2 protein levels normalized to β-actin. Bars represent mean ± sem. Statistical significance was assessed using Student’s t-test. *P<0.05, **P<0.01, ns, not significant (N=5). (C) The dendritic spines of hippocampal neurons were visualized by Golgi staining. Scale bar, 50 μm and 5 μm. (D) Dendritic spine density was quantified by ImageJ and assessed using Student’s t-test. **P<0.01 (N=7). (E) The mRNA levels of Shank1, Shank2, Shank3, and PSD-95 were quantified and normalized to β-actin. Bars represent mean ± sem. Statistical significance was assessed using Student’s t-test. *P < 0.05, **P < 0.01 (N = 6).
Figure 3. The influence of injecting AAV-shRTN4 into the rat hippocampus on the learning and memory behavior of rats exposed to HH. (A) Schematic for stereotaxic injection. AAV-shRTN4 or AAV-Scramble solution was injected into both sides of the hippocampus. The stereotaxic coordinates used were: AP = -3.8 mm, ML = ± 3.0 mm, DV = 3.2 mm relative to the bregma. (B) Representative western blot of Nogo-A and β-actin in rat hippocampus lysates. (C) The quantification of Nogo-A protein levels normalized to β-actin. Bars represent mean ± sem. Statistical significance was assessed using Student’s t-test. *P<0.05, ns, not significant (N=3). (D) Freezing time (%) in context test was measured after electric shock training and the freezing time (%) of Scramble HH and shRTN4 HH groups were analyzed. Statistical significance was assessed using one-way ANOVA followed by Tukey post hoc test. *P<0.05, ***P<0.001, ns, not significant (N=12). (E) Swimming distance to the platform, (F) primary escape latency, (G) time spent in the target quadrant, and (H) number of target entries in the probe testing phase. Bars represent mean ± sem. Statistical significance was assessed using one-way ANOVA followed by Tukey post hoc test. *P < 0.05, **P < 0.01, ***P < 0.001, ns, not significant (N = 10).
Figure 4. The influence of injecting AAV-shRTN4 into the rat hippocampus on hippocampal oxidative stress and dendritic spine density following exposure to HH. (A) Quantification of ROS levels, (B) MDA levels, (C) SOD activity, and (D) GSH/GSSG ratio in hippocampus homogenates after HH exposure. Bars represent mean ± sem. Statistical significance was assessed using one-way ANOVA followed by Tukey post hoc test. * P< 0.05, ***P < 0.001, ns, not significant (N = 6). (E-F) The dendritic spines of neurons in hippocampus were visualized and quantified. Scale bar, 5 μm. Statistical significance was assessed using one-way ANOVA followed by Tukey post hoc tests. **P < 0.05 (N = 7).
Figure 5. Nogo-A knockout improved HH-induced L&M damage. (A) Representative western blot images of Nogo-A and β-actin in the hippocampal homogenates of WT or Nogo-A-/- rats. (B) Freezing time (%) in context test was measured after electric shock training. Statistical significance was assessed using one-way ANOVA followed by Tukey post hoc tests. *P<0.05, ***P<0.001, ns, not significant (N=12). (C) Escape distance to the platform, (D) primary escape latency, (E) time spent in the target quadrant, and (F) number of target entries in the probe testing phase. (G) Representative swimming tracks in the MWM test. Data were analyzed using one-way ANOVA followed by Tukey post hoc tests. *P < 0.05, **P < 0.01, ***P < 0.001, ns, not significant (N = 10).
Figure 6. The influence of the knockdown of Nogo-A in SH-SY5Y cells on OGD/R-induced oxidative stress response. (A) Representative immunofluorescence images of Nogo-A in SH-SY5Y cells. (B) Representative western blot images of Nogo-A and β-actin in SH-SY5Y cells after ShRNA treatment. The protein levels were normalized to β-actin. Bars represent mean ± sem. Statistical significance was assessed using Student’s t-test. ***P < 0.001 (N = 4). (C) Representative flow cytometry images of Dihydroethidium (DHE) staining, and (D) quantification of intracellular ROS in SH-SY5Y cells after OGD/R with administration of LV-scramble or LV-ShRTN4. Bars represent mean ± sem. Statistical significance was assessed using one-way ANOVA followed by Tukey post hoc tests. ***P < 0.001 ns, not significant (N = 4). (E) Representative TEM images of the mitochondria in SH-SY5Y cells after OGD/R with administration of LV-scramble or LV-ShRTN4. Scale bars (500 nm) are indicated in images. (F) Quantification of the ratio of depolarization of the cell mitochondrial membrane potential. Bars represent mean ± sem. Statistical significance was assessed using one-way ANOVA followed by Tukey post hoc tests. *P < 0.05, ***P < 0.001, ns, not significant (N = 4).
Figure 7. The effects of NgR1 agonists on mitochondrial morphology and function in SH-SY5Y cells treated with shRTN4 under the OGD/R modeling. (A) Expression of NgR1 in SH-SY5Y cells was determined using western blot. Representative western blot images of NgR1 and β-actin in SH-SY5Y cells. (B) Quantification of intracellular ROS in Nogo-A Knockdown cells after OGD/R with the treatment of Nogo-P4 or Rtn-P4. Bars represent mean ± sem. Statistical significance was assessed using one-way ANOVA followed by Tukey post hoc tests. ***P<0.001, ns, not significant (N=4). (C) Representative TEM images of the mitochondria in SH-SY5Y cells treated with Nogo-P4 or Rtn-P4. Scale bars (500 nm) are indicated in the images. (D) Quantification of the ratio of depolarization of cell mitochondrial membrane potential. Bars represent mean ± sem. Scale bars assessed using one-way ANOVA followed by Tukey post hoc tests. *P < 0.05, **P < 0.01, ns, not significant (N = 4).
Figure 8. The effects of Nogo66 and NogoΔ20 antagonists on rat behavior after exposure to HH. (A) Freezing time (%) in the context test was measured after electric shock training. Statistical significance was assessed using one-way ANOVA followed by Tukey post hoc tests. **P<0.01, ns, not significant (N=12). (B) Swimming distance to the platform, (C) primary escape latency, (D) time spent in the target quadrant, and (E) number of target entries in the probe testing phase. (F) Representative swimming tracks in the MWM test. Statistical significance was analyzed using one-way ANOVA followed by Tukey post hoc tests. **P<0.01, ***P<0.001, ns, not significant (N=12).
-
[1] Wilson MH, Newman S, Imray CH. The cerebral effects of ascent to high altitudes. Lancet Neurol, 2009; 8, 175−91. doi: 10.1016/S1474-4422(09)70014-6 [2] Tremblay JC, Ainslie PN. Global and country-level estimates of human population at high altitude. Proc Natl Acad Sci USA, 2021; 118, e2102463118. doi: 10.1073/pnas.2102463118 [3] Richalet JP, Larmignat P, Poitrine E, et al. Physiological risk factors for severe high-altitude illness: a prospective cohort study. Am J Respir Crit Care Med, 2012; 185, 192−8. doi: 10.1164/rccm.201108-1396OC [4] Wakhloo D, Scharkowski F, Curto Y, et al. Functional hypoxia drives neuroplasticity and neurogenesis via brain erythropoietin. Nat Commun, 2020; 11, 1313. doi: 10.1038/s41467-020-15041-1 [5] Zhang ZA, Sun YF, Yuan ZY, et al. Insight into the effects of high-altitude hypoxic exposure on learning and memory. Oxid Med Cell Longev, 2022; 2022, 4163188. [6] Coimbra-Costa D, Alva N, Duran M, et al. Oxidative stress and apoptosis after acute respiratory hypoxia and reoxygenation in rat brain. Redox Biol, 2017; 12, 216−25. doi: 10.1016/j.redox.2017.02.014 [7] Iampietro M, Giovannetti T, Tarazi R. Hypoxia and inflammation in children with sickle cell disease: implications for hippocampal functioning and episodic memory. Neuropsychol Rev, 2014; 24, 252−65. doi: 10.1007/s11065-014-9259-4 [8] Pernet V, Schwab ME. The role of Nogo-A in axonal plasticity, regrowth and repair. Cell Tissue Res, 2012; 349, 97−104. doi: 10.1007/s00441-012-1432-6 [9] Chen MS, Huber AB, van der Haar ME, et al. Nogo-A is a myelin-associated neurite outgrowth inhibitor and an antigen for monoclonal antibody IN-1. Nature, 2000; 403, 434−9. doi: 10.1038/35000219 [10] Cao Y, Shumsky JS, Sabol MA, et al. Nogo-66 receptor antagonist peptide (NEP1-40) administration promotes functional recovery and axonal growth after lateral funiculus injury in the adult rat. Neurorehabil Neural Repair, 2008; 22, 262−78. doi: 10.1177/1545968307308550 [11] Schweigreiter R. The natural history of the myelin-derived nerve growth inhibitor Nogo-A. Neuron Glia Biol, 2008; 4, 83−9. doi: 10.1017/S1740925X09990147 [12] Zemmar A, Weinmann O, Kellner Y, et al. Neutralization of Nogo-A enhances synaptic plasticity in the rodent motor cortex and improves motor learning in vivo. J Neurosci, 2014; 34, 8685−98. doi: 10.1523/JNEUROSCI.3817-13.2014 [13] Vajda F, Jordi N, Dalkara D, et al. Cell type-specific Nogo-A gene ablation promotes axonal regeneration in the injured adult optic nerve. Cell Death Differ, 2015; 22, 323−35. doi: 10.1038/cdd.2014.147 [14] Seiler S, Widmer HR. Nogo-A and its functions beyond axonal inhibition: the controversial role of Nogo-A in Parkinson's disease. Neural Regen Res, 2015; 10, 1223−4. doi: 10.4103/1673-5374.162749 [15] Fang YQ, Yao LM, Li CH, et al. The blockage of the Nogo/NgR signal pathway in microglia alleviates the formation of Aβ plaques and tau phosphorylation in APP/PS1 transgenic mice. J Neuroinflammation, 2016; 13, 56. doi: 10.1186/s12974-016-0522-x [16] Ineichen BV, Plattner PS, Good N, et al. Nogo-A antibodies for progressive multiple sclerosis. CNS Drugs, 2017; 31, 187−98. doi: 10.1007/s40263-017-0407-2 [17] Wang YY, Han N, Hong DJ, et al. Nogo-A aggravates oxidative damage in oligodendrocytes. Neural Regen Res, 2021; 16, 179−85. doi: 10.4103/1673-5374.286979 [18] Gil V, Nicolas O, Mingorance A, et al. Nogo-A expression in the human hippocampus in normal aging and in Alzheimer disease. J Neuropathol Exp Neurol, 2006; 65, 433−44. doi: 10.1097/01.jnen.0000222894.59293.98 [19] Gil V, Nicolas O, Mingorance A, et al. Nogo-A expression in the human hippocampus in normal aging and in Alzheimer disease. J Neuropathol Exp Neurol, 2006; 65, 433-44. [20] Zemmar A, Weinmann O, Kellner Y, et al. Neutralization of Nogo-A enhances synaptic plasticity in the rodent motor cortex and improves motor learning in vivo. J Neurosci, 2014; 34, 8685-98. [21] Wang H, Yao YJ, Jiang XN, et al. Expression of Nogo-A and NgR in the developing rat brain after hypoxia-ischemia. Brain Res, 2006; 1114, 212−20. doi: 10.1016/j.brainres.2006.07.056 [22] Wiessner C, Bareyre FM, Allegrini PR, et al. Anti-Nogo-A antibody infusion 24 hours after experimental stroke improved behavioral outcome and corticospinal plasticity in normotensive and spontaneously hypertensive rats. J Cereb Blood Flow Metab, 2003; 23, 154−65. doi: 10.1097/01.WCB.0000040400.30600.AF [23] Dosek A, Ohno H, Acs Z, et al. High altitude and oxidative stress. Respir Physiol Neurobiol, 2007; 158, 128−31. doi: 10.1016/j.resp.2007.03.013 [24] Zagrebelsky M, Schweigreiter R, Bandtlow CE, et al. Nogo-A stabilizes the architecture of hippocampal neurons. J Neurosci, 2010; 30, 13220−34. doi: 10.1523/JNEUROSCI.1044-10.2010 [25] Vallejo D, Codocedo JF, Inestrosa NC. Posttranslational modifications regulate the postsynaptic localization of PSD-95. Mol Neurobiol, 2017; 54, 1759−76. doi: 10.1007/s12035-016-9745-1 [26] Naisbitt S, Kim E, Tu JC, et al. Shank, a novel family of postsynaptic density proteins that binds to the NMDA receptor/PSD-95/GKAP complex and cortactin. Neuron, 1999; 23, 569−82. doi: 10.1016/S0896-6273(00)80809-0 [27] Xie LH, Fefelova N, Pamarthi SH, et al. Molecular mechanisms of ferroptosis and relevance to cardiovascular disease. Cells, 2022; 11, 2726. doi: 10.3390/cells11172726 [28] Szabò I, Leanza L, Gulbins E, et al. Physiology of potassium channels in the inner membrane of mitochondria. Pflügers Arch-Eur J Physiol, 2012; 463, 231−46. [29] Sui YP, Zhang XX, Lu JL, et al. New insights into the roles of Nogo-A in CNS biology and diseases. Neurochem Res, 2015; 40, 1767−85. doi: 10.1007/s11064-015-1671-5 [30] Zhang G, Zhou SM, Yuan C, et al. The effects of short-term and long-term exposure to a high altitude hypoxic environment on neurobehavioral function. High Alt Med Biol, 2013; 14, 338−41. doi: 10.1089/ham.2012.1091 [31] Mairer K, Wille M, Bucher T, et al. Prevalence of acute mountain sickness in the Eastern Alps. High Alt Med Biol, 2009; 10, 239−45. doi: 10.1089/ham.2008.1091 [32] Maggiorini M, Bühler B, Walter M, et al. Prevalence of acute mountain sickness in the Swiss Alps. BMJ, 1990; 301, 853−5. doi: 10.1136/bmj.301.6756.853 [33] Delekate A, Zagrebelsky M, Kramer S, et al. NogoA restricts synaptic plasticity in the adult hippocampus on a fast time scale. Proc Natl Acad Sci USA, 2011; 108, 2569−74. doi: 10.1073/pnas.1013322108 [34] Tang BL. Nogo-A and the regulation of neurotransmitter receptors. Neural Regen Res, 2020; 15, 2037−8. doi: 10.4103/1673-5374.282250 [35] Wills ZP, Mandel-Brehm C, Mardinly AR, et al. The nogo receptor family restricts synapse number in the developing hippocampus. Neuron, 2012; 73, 466−81. doi: 10.1016/j.neuron.2011.11.029 [36] Murthy R, Kim J, Sun XK, et al. Post-transcriptional regulation of GABAB receptor and GIRK1 channels by Nogo receptor 1. Mol Brain, 2013; 6, 30. doi: 10.1186/1756-6606-6-30 [37] Salim S. Oxidative stress and the central nervous system. J Pharmacol Exp Ther, 2017; 360, 201−5. doi: 10.1124/jpet.116.237503 [38] Chamberlain KA, Sheng ZH. Mechanisms for the maintenance and regulation of axonal energy supply. J Neurosci Res, 2019; 97, 897−913. doi: 10.1002/jnr.24411 [39] Devine MJ, Kittler JT. Mitochondria at the neuronal presynapse in health and disease. Nat Rev Neurosci, 2018; 19, 63−80. [40] Sharma C, Kim S, Nam Y, et al. Mitochondrial dysfunction as a driver of cognitive impairment in Alzheimer’s disease. Int J Mol Sci, 2021; 22, 4850. doi: 10.3390/ijms22094850 [41] Auerbach JM, Segal M. Peroxide modulation of slow onset potentiation in rat hippocampus. J Neurosci, 1997; 17, 8695−701. doi: 10.1523/JNEUROSCI.17-22-08695.1997