75
Views
0
CrossRef citations to date
0
Altmetric
Original Research

Astrocyte HIF-2α supports learning in a passive avoidance paradigm under hypoxic stress

, , , , , , , , , , , , & show all
Pages 35-56 | Published online: 08 Nov 2018

References

  • Pittman RN. Regulation of Tissue Oxygenation. San Rafael, CA: Morgan & Claypool Life Sciences; 2011.
  • Bruce AS, Aloia MS, Ancoli-Israel S. Neuropsychological effects of hypoxia in medical disorders. In: Grant I, Adams KM, editors. Neuropsychological Assessment of Neuropsychiatric and Neuromedical Disorders. 3rd ed. Oxford, New York: Oxford University Press; 2009:336–349.
  • Cannon JA, Moffitt P, Perez-Moreno AC, et al. Cognitive Impairment and Heart Failure: Systematic Review and Meta-Analysis. J Card Fail. 2017;23:464–475.
  • Yohannes AM, Chen W, Moga AM, Leroi I, Connolly MJ. Cognitive Impairment in Chronic Obstructive Pulmonary Disease and Chronic Heart Failure: A Systematic Review and Meta-analysis of Observational Studies. J Am Med Dir Assoc. 2017;18(5):451.e1–451.e11.
  • Schega L, Peter B, Törpel A, Mutschler H, Isermann B, Hamacher D. Effects of intermittent hypoxia on cognitive performance and quality of life in elderly adults: a pilot study. Gerontology. 2013;59(4):316–323.
  • Qiu C, Fratiglioni L. A major role for cardiovascular burden in age-related cognitive decline. Nat Rev Cardiol. 2015;12(5):267–277.
  • Popa-Wagner A, Buga A-M, Popescu B, Muresanu D. Vascular cognitive impairment, dementia, aging and energy demand. A vicious cycle. J Neural Transm. 2015;122(S1):47–54.
  • Lahousse L, Tiemeier H, Ikram MA, Brusselle GG. Chronic obstructive pulmonary disease and cerebrovascular disease: A comprehensive review. Respir Med. 2015;109(11):1371–1380.
  • Daulatzai MA. Evidence of neurodegeneration in obstructive sleep apnea: Relationship between obstructive sleep apnea and cognitive dysfunction in the elderly. J Neurosci Res. 2015;93(12):1778–1794.
  • Bressler J, Knopman DS, Sharrett AR, et al. Incident Heart Failure and Cognitive Decline: The Atherosclerosis Risk in Communities Study. J Card Fail. 2017;23(1):47–55.
  • Suzuki H, Matsumoto Y, Ota H, et al. Hippocampal Blood Flow Abnormality Associated With Depressive Symptoms and Cognitive Impairment in Patients With Chronic Heart Failure. Circ J. 2016;80(8):1773–1780.
  • Kurella Tamura M, Vittinghoff E, Yang J, et al. Anemia and risk for cognitive decline in chronic kidney disease. BMC Nephrol. 2016;17(1):13.
  • Dlugaj M, Winkler A, Weimar C, et al. Anemia and Mild Cognitive Impairment in the German General Population. J Alzheimers Dis. 2016;49:1031–1042.
  • Schneider ALC, Jonassaint C, Sharrett AR, et al. Hemoglobin, Anemia, and Cognitive Function: The Atherosclerosis Risk in Communities Study. J Gerontol A Biol Sci Med Sci. 2016;71(6):772–779.
  • Weiskopf RB, Kramer JH, Viele M, et al. Acute Severe Isovolemic Anemia Impairs Cognitive Function and Memory in Humans. Anesthesiology. 2000;92(6):1646–1652.
  • Weiskopf RB, Feiner J, Hopf HW, et al. Oxygen Reverses Deficits of Cognitive Function and Memory and Increased Heart Rate Induced by Acute Severe Isovolemic Anemia. Anesthesiology. 2002;96(4):871–877.
  • Hornbein TF. The high-altitude brain. J Exp Biol. 2001;204(Pt 18):3129–3132.
  • Wilson MH, Newman S, Imray CH. The cerebral effects of ascent to high altitudes. Lancet Neurol. 2009;8(2):175–191.
  • Neuhaus C, Hinkelbein J. Cognitive responses to hypobaric hypoxia: implications for aviation training. Psychol Res Behav Manag. 2014;7:297–302.
  • Malle C, Quinette P, Laisney M, et al. Working Memory Impairment in Pilots Exposed to Acute Hypobaric Hypoxia. Aviat Space Environ Med. 2013;84(8):773–779.
  • Tran PV, Carlson ES, Fretham SJB, Georgieff MK. Early-Life Iron Deficiency Anemia Alters Neurotrophic Factor Expression and Hippocampal Neuron Differentiation in Male Rats. J Nutr. 2008;138(12):2495–2501.
  • Hennigan A, Callaghan CK, Kealy J, Rouine J, Kelly Áine M.. Deficits in LTP and recognition memory in the genetically hypertensive rat are associated with decreased expression of neurotrophic factors and their receptors in the dentate gyrus. Behav Brain Res. 2009;197(2):371–377.
  • Jänicke B, Schulze G. Influence of normobaric hypoxia on learning capacity of different aged rats. Neurobiol Aging. 1987;8(6):495–500.
  • Li M, Bertout JA, Ratcliffe SJ, Eckenhoff MF, Simon MC, Floyd TF. Acute Anemia Elicits Cognitive Dysfunction and Evidence of Cerebral Cellular Hypoxia in Older Rats with Systemic Hypertension. Anesthesiology. 2010;113(4):845–858.
  • Gozal D, Row BW, Kheirandish L, et al. Increased susceptibility to intermittent hypoxia in aging rats: changes in proteasomal activity, neuronal apoptosis and spatial function. J Neurochem. 2003;86(6):1545–1552.
  • Ingraham JP, Forbes ME, Riddle DR, Sonntag WE. Aging Reduces Hypoxia-Induced Microvascular Growth in the Rodent Hippocampus. J Gerontol A Biol Sci Med Sci. 2008;63(1):12–20.
  • Dunwiddie TV. Age-related differences in the in vitro rat hippocampus. Development of inhibition and the effects of hypoxia. Dev Neurosci. 1981;4:165–175.
  • Larson J, Drew KL, Folkow LP, Milton SL, Park TJ. No oxygen? No problem! Intrinsic brain tolerance to hypoxia in vertebrates. J Exp Biol. 2014;217(7):1024–1039.
  • Wang GL, Jiang BH, Rue EA, Semenza GL. Hypoxia-inducible factor 1 is a basic-helix-loop-helix-PAS heterodimer regulated by cellular O2 tension. Proc Natl Acad Sci U S A. 1995;92(12):5510–5514.
  • Sharp FR, Bergeron M, Bernaudin M. Hypoxia-inducible factor in brain. Adv Exp Med Biol. 2001;502:273–291.
  • Lahiri S, Roy A, Baby SM, Hoshi T, Semenza GL, Prabhakar NR. Oxygen sensing in the body. Prog Biophys Mol Biol. 2006;91(3):249–286.
  • Kaelin WG, Ratcliffe PJ. Oxygen Sensing by Metazoans: The Central Role of the HIF Hydroxylase Pathway. Mol Cell. 2008;30(4):393–402.
  • Prabhakar NR, Semenza GL. Oxygen Sensing and Homeostasis. Physiology (Bethesda). 2015;30:340–348.
  • Mclaren AT, Mazer CD, Zhang H, Liu E, Mok L, Hare GM. A potential role for inducible nitric oxide synthase in the cerebral response to acute hemodilution. Can J Anaesth. 2009;56(7):502–509.
  • Powell FL, Kim BC, Johnson SR, Fu Z. Oxygen sensing in the brain-invited article. Adv Exp Med Biol. 2009;648:369–376.
  • Lamanna JC. Hypoxia in the central nervous system. Essays Biochem. 2007;43:138–152.
  • Knabe W, Knerlich F, Washausen S, et al. Expression patterns of erythropoietin and its receptor in the developing midbrain. Anat Embryol. 2004;207(6):503–512.
  • Schodel J, Oikonomopoulos S, Ragoussis J, Pugh CW, Ratcliffe PJ, Mole DR. High-resolution genome-wide mapping of HIF-binding sites by ChIP-seq. Blood. 2011;117(23):e207–e217.
  • Wenger RH. Mammalian oxygen sensing, signalling and gene regulation. J Exp Biol. 2000;203(Pt 8):1253–1263.
  • Semenza GL. Oxygen Sensing, Hypoxia-Inducible Factors, and Disease Pathophysiology. Annu Rev Pathol. 2014;9(1):47–71.
  • Rivard A, Berthou-Soulie L, Principe N, et al. Age-dependent defect in vascular endothelial growth factor expression is associated with reduced hypoxia-inducible factor 1 activity. J Biol Chem. 2000;275(38):29643–29647.
  • Chavez JC, Lamanna JC. Hypoxia-inducible factor-1alpha accumulation in the rat brain in response to hypoxia and ischemia is attenuated during aging. Adv Exp Med Biol. 2003;510:337–341.
  • Giulio CD, Bianchi G, Cacchio M, et al. Oxygen and life span: chronic hypoxia as a model for studying HIF-1α, VEGF and NOS during aging. Respir Physiol Neurobiol. 2005;147(1):31–38.
  • Bosch-Marce M, Okuyama H, Wesley JB, et al. Effects of Aging and Hypoxia-Inducible Factor-1 Activity on Angiogenic Cell Mobilization and Recovery of Perfusion After Limb Ischemia. Circ Res. 2007;101(12):1310–1318.
  • Katschinski D. Is there a molecular connection between hypoxia and aging? Exp Gerontol. 2006;41(5):482–484.
  • Ndubuizu OI, Chavez JC, Lamanna JC. Increased prolyl 4-hydroxylase expression and differential regulation of hypoxia-inducible factors in the aged rat brain. Am J Physiol Regul Integr Comp Physiol. 2009;297(1):R158–R165.
  • Rapino C, Bianchi G, di Giulio C, et al. HIF-1alpha cytoplasmic accumulation is associated with cell death in old rat cerebral cortex exposed to intermittent hypoxia. Aging Cell. 2005;4(4):177–185.
  • Frenkel-Denkberg G, Gershon D, Levy AP. The function of hypoxia-inducible factor 1 (HIF-1) is impaired in senescent mice. FEBS Lett. 1999;462(3):341–344.
  • Kim H, Lee D-K, Choi J-W, Kim J-S, Park SC, Youn H-D. Analysis of the effect of aging on the response to hypoxia by cDNA microarray. Mech Ageing Dev. 2003;124(8-9):941–949.
  • Benderro GF, Lamanna JC. Hypoxia-induced angiogenesis is delayed in aging mouse brain. Brain Res. 2011;1389:50–60.
  • Rabie T, Kunze R, Marti HH. Impaired hypoxic response in senescent mouse brain. Int J Dev Neurosci. 2011;29(6):655–661.
  • Anderson J, Sandhir R, Hamilton ES, Berman NEJ. Impaired Expression of Neuroprotective Molecules in the HIF-1α Pathway following Traumatic Brain Injury in Aged Mice. J Neurotrauma. 2009;26(9):1557–1566.
  • Chavez JC, Baranova O, Lin J, Pichiule P. The transcriptional activator hypoxia inducible factor 2 (HIF-2/EPAS-1) regulates the oxygen-dependent expression of erythropoietin in cortical astrocytes. J Neurosci. 2006;26(37):9471–9481.
  • Schousboe A, Sickmann HM, Bak LK, et al. Neuron-glia interactions in glutamatergic neurotransmission: Roles of oxidative and glycolytic adenosine triphosphate as energy source. J Neurosci Res. 2011;89(12):1926–1934.
  • Dienel GA. Astrocytic energetics during excitatory neurotransmission: What are contributions of glutamate oxidation and glycolysis? Neurochem Int. 2013;63(4):244–258.
  • Harris JJ, Jolivet R, Attwell D. Synaptic Energy Use and Supply. Neuron. 2012;75(5):762–777.
  • Lu B, Nagappan G, Lu Y. BDNF and synaptic plasticity, cognitive function, and dysfunction. In: Lewin GR, Carter BD, editors. Neurotrophic Factors. Berlin: Springer. 2014:223–250.
  • Lum JJ, Bui T, Gruber M, et al. The transcription factor HIF-1 plays a critical role in the growth factor-dependent regulation of both aerobic and anaerobic glycolysis. Genes Dev. 2007;21(9):1037–1049.
  • Jelkmann W. Regulation of erythropoietin production. J Physiol. 2011;589(6):1251–1258.
  • Nakazawa MS, Keith B, Simon MC. Oxygen availability and metabolic adaptations. Nat Rev Cancer. 2016;16(10):663–673.
  • Sardinha VM, Guerra-Gomes S, Caetano I, et al. Astrocytic signaling supports hippocampal-prefrontal theta synchronization and cognitive function. Glia. 2017;65(12):1944–1960.
  • Holthoff K, Witte OW. Directed spatial potassium redistribution in rat neocortex. Glia. 2000;29(3):288–292.
  • Kofuji P, Newman EA. Potassium buffering in the central nervous system. Neuroscience. 2004;129(4):1043–1054.
  • Ota Y, Zanetti AT, Hallock RM. The Role of Astrocytes in the Regulation of Synaptic Plasticity and Memory Formation. Neural Plast. 2013;2013(1):11–11.
  • Allen NJ. Astrocyte Regulation of Synaptic Behavior. Annu Rev Cell Dev Biol. 2014;30(1):439–463.
  • Christopherson KS, Ullian EM, Stokes CCA, et al. Thrombospondins Are Astrocyte-Secreted Proteins that Promote CNS Synaptogenesis. Cell. 2005;120(3):421–433.
  • Risher WC, Eroglu C. Thrombospondins as key regulators of synaptogenesis in the central nervous system. Matrix Biology. 2012;31(3):170–177.
  • Pannasch U, Vargová L, Reingruber J, et al. Astroglial networks scale synaptic activity and plasticity. Proc Natl Acad Sci U S A. 2011;108(20):8467–8472.
  • Stogsdill JA, Eroglu C. The interplay between neurons and glia in synapse development and plasticity. Curr Opin Neurobiol. 2017;42:1–8.
  • Gruber M, Hu CJ, Johnson RS, Brown EJ, Keith B, Simon MC. Acute postnatal ablation of Hif-2alpha results in anemia. Proc Natl Acad Sci U S A. 2007;104(7):2301–2306.
  • Weidemann A, Kerdiles YM, Knaup KX, et al. The glial cell response is an essential component of hypoxia-induced erythropoiesis in mice. J Clin Invest. 2009;119:3373–3383.
  • Liu R, Suzuki A, Guo Z, Mizuno Y, Urabe T. Intrinsic and extrinsic erythropoietin enhances neuroprotection against ischemia and reperfusion injury in vitro. J Neurochem. 2006;96(4):1101–1110.
  • Petzold GC, Murthy VN. Role of Astrocytes in Neurovascular Coupling. Neuron. 2011;71(5):782–797.
  • Tarantini S, Tran CHT, Gordon GR, Ungvari Z, Csiszar A. Impaired neurovascular coupling in aging and Alzheimer’s disease: Contribution of astrocyte dysfunction and endothelial impairment to cognitive decline. Exp Gerontol. 2017;94:52–58.
  • Wang M, He Y, Sejnowski TJ, Yu X. Brain-state dependent astrocytic Ca2+ signals are coupled to both positive and negative BOLD-fMRI signals. Proc Natl Acad Sci U S A. 2018;115(7):E1647–E1656.
  • Bolaños JP, Cidad P, Garcı́a-Nogales P, Delgado-Esteban M, Fernández E, Almeida A. Regulation of glucose metabolism by nitrosative stress in neural cells. Mol Aspects Med. 2004;25(1-2):61–73.
  • Ryan HE, Poloni M, Mcnulty W, et al. Hypoxia-inducible factor-1alpha is a positive factor in solid tumor growth. Cancer Res. 2000;60:4010–4015.
  • Sakr HF. Abbas AM and El Samanoudy AZ. Effect of vitamin E on cerebral cortical oxidative stress and brain-derived neurotrophic factor gene expression induced by hypoxia and exercise in rats. J Physiol Pharmacol. 2015;66:191–202.
  • Bell M, Thake CD, Wolff CB. Effect of inspiration of 12% O2 (Balance N2) on cardiac output, respiration, oxygen saturation, and oxygen delivery. In: LaManna JC, editor. Oxygen Transport to Tissue New York. Vol. (XXXII). Berlin, Germany: Springer; 2011:327–332.
  • Group TL-TOTTR. A Randomized Trial of Long-Term Oxygen for COPD with Moderate Desaturation. New England Journal of Medicine. 2016;375:1617–1627.
  • Kim C-H. The effects of hypoxia and exercise on physiological and cognitive performance. 2011. Available from: https://etd.ohiolink.edu/pg_10?0::NO:10:P10_ACCESSION_NUM:kent1311015088. Accessed September 05, 2018.
  • Gray A, Goodacre S, Newby DE, Masson M, Sampson F, Nicholl J. Noninvasive Ventilation in Acute Cardiogenic Pulmonary Edema. N Engl J Med Overseas Ed. 2008;359(2):142–151.
  • Forsythe JA, Jiang BH, Iyer NV, et al. Activation of vascular endothelial growth factor gene transcription by hypoxia-inducible factor 1. Mol Cell Biol. 1996;16(9):4604–4613.
  • Cable GG. In-flight hypoxia incidents in military aircraft: causes and implications for training. Aviat Space Environ Med. 2003;74:169–172.
  • Stroka DM, Burkhardt T, Desbaillets I, et al. HIF-1 is expressed in normoxic tissue and displays an organ-specific regulation under systemic hypoxia. Faseb J. 2001;15(13):2445–2453.
  • Vogelmeier CF, Criner GJ, Martinez FJ, et al. Global Strategy for the Diagnosis, Management, and Prevention of Chronic Obstructive Lung Disease 2017 Report. GOLD Executive Summary. Am J Respir Crit Care Med. 2017;195(5):557–582.
  • Cranston JM, Crockett A, Moss J, Alpers JH, Cochrane Airways Group. Domiciliary oxygen for chronic obstructive pulmonary disease. Cochrane Database Syst Rev. 2005;14(5).
  • Burman MA, Simmons CA, Hughes M, Lei L. Developing and validating trace fear conditioning protocols in C57BL/6 mice. J Neurosci Methods. 2014;222:111–117.
  • Mccarthy KD, de Vellis J. Preparation of separate astroglial and oligodendroglial cell cultures from rat cerebral tissue. J Cell Biol. 1980;85(3):890–902.
  • Olney JW, Tenkova T, Dikranian K, Qin YQ, Labruyere J, Ikonomidou C. Ethanol-induced apoptotic neurodegeneration in the developing C57BL/6 mouse brain. Brain Res Dev Brain Res. 2002;133(2):115–126.
  • Lamberty Y, Gower AJ. Age-related changes in spontaneous behavior and learning in NMRI mice from maturity to middle age. Physiol Behav. 1990;47(6):1137–1144.
  • Jahn-Eimermacher A, Lasarzik I, Raber J. Statistical analysis of latency outcomes in behavioral experiments. Behav Brain Res. 2011;221(1):271–275.
  • Tomita S, Ueno M, Sakamoto M, et al. Defective Brain Development in Mice Lacking the Hif-1 Gene in Neural Cells. Mol Cell Biol. 2003;23(19):6739–6749.
  • Cy K, Tsai MY, Tseng WF, et al. Integration of CNS survival and differentiation by HIF2alpha. Cell Death Differ. 2011;18:1757–1770.
  • Suzuki A, Stern SA, Bozdagi O, et al. Astrocyte-Neuron Lactate Transport Is Required for Long-Term Memory Formation. Cell. 2011;144(5):810–823.
  • Adamcio B, Sargin D, Stradomska A, et al. Erythropoietin enhances hippocampal long-term potentiation and memory. BMC Biol. 2008;6(1):37.
  • Ögren SO, Kuteeva E, Elvander-Tottie E, Hökfelt T. Neuropeptides in learning and memory processes with focus on galanin. Eur J Pharmacol. 2010;626(1):9–17.
  • Detrait ER, Hanon E, Dardenne B, Lamberty Y. The inhibitory avoidance test optimized for discovery of cognitive enhancers. Behav Res Methods. 2009;41(3):805–811.
  • Antunes M, Biala G. The novel object recognition memory: neurobiology, test procedure, and its modifications. Cogn Process. 2012;13(2):93–110.
  • Krypotos A-M, Effting M, Kindt M, Beckers T. Avoidance learning: a review of theoretical models and recent developments. Front Behav Neurosci. 2015;9:189.
  • Pinho J, Vale R, Batalha VL, et al. Enhanced LTP in aged rats: Detrimental or compensatory? Neuropharmacology. 2017;114:12–19.
  • Costenla AR, de Mendonça A, Ribeiro JA. Adenosine modulates synaptic plasticity in hippocampal slices from aged rats. Brain Res. 1999; 851(1-2):228–234.
  • Kumar A, Foster TC. Enhanced Long-Term Potentiation During Aging Is Masked by Processes Involving Intracellular Calcium Stores. J Neurophysiol. 2004;91(6):2437–2444.
  • Huang Y-Y, Kandel ER. Age-related enhancement of a protein synthesis-dependent late phase of LTP induced by low frequency paired-pulse stimulation in hippocampus. Learn Mem. 2006;13(3):298–306.
  • Diógenes MJ, Fernandes CC, Sebastião AM, Ribeiro JA. Activation of adenosine A2A receptor facilitates brain-derived neurotrophic factor modulation of synaptic transmission in hippocampal slices. J Neurosci. 2004;24(12):2905–2913.
  • Kuschel A, Simon P, Tug S. Functional regulation of HIF-1α under normoxia-is there more than post-translational regulation? J Cell Physiol. 2012;227(2):514–524.
  • Pagé EL, Chan DA, Giaccia AJ, Levine M, Richard DE. Hypoxia-inducible factor-1alpha stabilization in nonhypoxic conditions: role of oxidation and intracellular ascorbate depletion. Mol Biol Cell. 2008;19(1):86–94.
  • Baek SH, Kim KI. Regulation of HIF-1α stability by lysine methylation. BMB Rep. 2016;49(5):245–246.
  • Mamczur P, Borsuk B, Paszko J, et al. Astrocyte-neuron crosstalk regulates the expression and subcellular localization of carbohydrate metabolism enzymes. Glia. 2015;63(2):328–340.
  • Tsuboi I, Yamashita T, Nagano M, Kimura K, To’a Salazar G, Ohneda O. Impaired expression of HIF-2α induces compensatory expression of HIF-1α for the recovery from anemia. J Cell Physiol. 2015;230(7):1534–1548.
  • Jha MK, Kim JH, Song GJ, et al. Functional dissection of astrocyte-secreted proteins: Implications in brain health and diseases. Prog Neurobiol. 2018;162:37-69.
  • Zhao J, du F, Shen G, Zheng F, Xu B. The role of hypoxia-inducible factor-2 in digestive system cancers. Cell Death Dis. 2015;6:e1600.
  • Holmquist-Mengelbier L, Fredlund E, Löfstedt T, et al. Recruitment of HIF-1alpha and HIF-2alpha to common target genes is differentially regulated in neuroblastoma: HIF-2alpha promotes an aggressive phenotype. Cancer Cell. 2006;10(5):413–423.
  • Filosa JA, Blanco VM. Neurovascular coupling in the mammalian brain. Exp Physiol. 2007;92(4):641–646.
  • Girouard H, Iadecola C. Neurovascular coupling in the normal brain and in hypertension, stroke, and Alzheimer disease. J Appl Physiol. 2006;100(1):328–335.
  • Iadecola C. The Neurovascular Unit Coming of Age: A Journey through Neurovascular Coupling in Health and Disease. Neuron. 2017;96(1):17–42.
  • Perea G, Navarrete M, Araque A. Tripartite synapses: astrocytes process and control synaptic information. Trends Neurosci. 2009;32(8):421–431.
  • Koehler RC, Roman RJ, Harder DR. Astrocytes and the regulation of cerebral blood flow. Trends Neurosci. 2009;32(3):160–169.
  • Marina N, Turovsky E, Christie IN, et al. Brain metabolic sensing and metabolic signaling at the level of an astrocyte. Glia. 2018;66(6):1185-1199.
  • Vallon M, Chang J, Zhang H, Kuo CJ. Developmental and pathological angiogenesis in the central nervous system. Cell Mol Life Sci. 2014;71(18):3489–3506.
  • Jung F, Palmer LA, Zhou N, Johns RA. Hypoxic regulation of inducible nitric oxide synthase via hypoxia inducible factor-1 in cardiac myocytes. Circ Res. 2000;86(3):319–325.
  • Semenza GL, Nejfelt MK, Chi SM, Antonarakis SE. Hypoxia-inducible nuclear factors bind to an enhancer element located 3’ to the human erythropoietin gene. Proceedings of the National Academy of Sciences. 1991;88(13):5680–5684.
  • Esmaeili Tazangi P, Moosavi SM, Shabani M, Haghani M. Erythropoietin improves synaptic plasticity and memory deficits by decrease of the neurotransmitter release probability in the rat model of Alzheimer’s disease. Pharmacol Biochem Behav. 2015;130:15–21.
  • Dayyat EA, Zhang SX, Wang Y, Cheng ZJ, Gozal D. Exogenous erythropoietin administration attenuates intermittent hypoxia-induced cognitive deficits in a murine model of sleep apnea. BMC Neurosci. 2012;13:77.
  • Al-Qahtani JM, Abdel-Wahab BA, Abd El-Aziz SM. Long-term moderate dose exogenous erythropoietin treatment protects from intermittent hypoxia-induced spatial learning deficits and hippocampal oxidative stress in young rats. Neurochem Res. 2014;39(1):161–171.
  • Viviani B, Bartesaghi S, Corsini E, et al. Erythropoietin protects primary hippocampal neurons increasing the expression of brain-derived neurotrophic factor. J Neurochem. 2005;93(2):412–421.
  • Dzietko M, Felderhoff-Mueser U, Sifringer M, et al. Erythropoietin protects the developing brain against N-methyl-D-aspartate receptor antagonist neurotoxicity. Neurobiol Dis. 2004;15(2):177–187.
  • Mahmood A, Lu D, Qu C, et al. Treatment of traumatic brain injury in rats with erythropoietin and carbamylated erythropoietin. J Neurosurg. 2007;107(2):392–397.
  • Sönmez A, Kabakçi B, Vardar E, et al. Erythropoietin attenuates neuronal injury and potentiates the expression of pCREB in anterior horn after transient spinal cord ischemia in rats. Surg Neurol. 2007;68(3):297–303.
  • Zhong J, Zhang T, Bloch LM. Dendritic mRNAs encode diversified functionalities in hippocampal pyramidal neurons. BMC Neurosci. 2006;7:17.
  • Haase VH. Regulation of erythropoiesis by hypoxia-inducible factors. Blood Rev. 2013;27(1):41–53.
  • Kapitsinou PP, Liu Q, Unger TL, et al. Hepatic HIF-2 regulates erythropoietic responses to hypoxia in renal anemia. Blood. 2010;116(16):3039–3048.
  • Yeo EJ, Cho YS, Kim MS, Park JW. Contribution of HIF-1alpha or HIF-2alpha to erythropoietin expression: in vivo evidence based on chromatin immunoprecipitation. Ann Hematol. 2008;87(1):11–17.
  • Uniacke J, Holterman CE, Lachance G, et al. An oxygen-regulated switch in the protein synthesis machinery. Nature. 2012;486(7401):126–129.
  • Sakers K, Lake AM, Khazanchi R, et al. Astrocytes locally translate transcripts in their peripheral processes. Proc Natl Acad Sci U S A. 2017;114(19):E3830–E3838.
  • Jessen KR, Mirsky R. Nonmyelin-forming Schwann cells coexpress surface proteins and intermediate filaments not found in myelin-forming cells: a study of Ran-2, A5E3 antigen and glial fibrillary acidic protein. J Neurocytol. 1984;13(6):923–934.
  • Sarthy PV, Fu M. Transcriptional activation of an intermediate filament protein gene in mice with retinal dystrophy. DNA. 1989;8(6):437–446.
  • Feinstein DL, Weinmaster GA, Milner RJ. Isolation of cDNA clones encoding rat glial fibrillary acidic protein: expression in astrocytes and in Schwann cells. J Neurosci Res. 1992;32(1):1–14.
  • Doetsch F. The glial identity of neural stem cells. Nat Neurosci. 2003;6(11):1127–1134.
  • Casper KB, Jones K, Mccarthy KD. Characterization of astrocyte-specific conditional knockouts. Genesis. 2007;45(5):292–299.
  • Chow LM, Zhang J, Baker SJ. Inducible Cre recombinase activity in mouse mature astrocytes and adult neural precursor cells. Transgenic Res. 2008;17(5):919–928.
  • Bovolenta P, Liem RK, Mason CA. Glial filament protein expression in astroglia in the mouse visual pathway. Brain Res. 1987;430(1):113–126.
  • Bajenaru ML, Zhu Y, Hedrick NM, Donahoe J, Parada LF, Gutmann DH. Astrocyte-specific inactivation of the neurofibromatosis 1 gene (NF1) is insufficient for astrocytoma formation. Mol Cell Biol. 2002;22(14):5100–5113.
  • Lee Y, Messing A, Su M, Brenner M. GFAP promoter elements required for region-specific and astrocyte-specific expression. Glia. 2008;56(5):481–493.
  • Hirrlinger PG, Scheller A, Braun C, Hirrlinger J, Kirchhoff F. Temporal control of gene recombination in astrocytes by transgenic expression of the tamoxifen-inducible DNA recombinase variant CreERT2. Glia. 2006;54(1):11–20.