Propofol directly increases tau phosphorylation

Robert A Whittington, László Virág, François Marcouiller, Marie-Amélie Papon, Noura B El Khoury, Carl Julien, Françoise Morin, Charles W Emala, Emmanuel Planel, Robert A Whittington, László Virág, François Marcouiller, Marie-Amélie Papon, Noura B El Khoury, Carl Julien, Françoise Morin, Charles W Emala, Emmanuel Planel

Abstract

In Alzheimer's disease (AD) and other tauopathies, the microtubule-associated protein tau can undergo aberrant hyperphosphorylation potentially leading to the development of neurofibrillary pathology. Anesthetics have been previously shown to induce tau hyperphosphorylation through a mechanism involving hypothermia-induced inhibition of protein phosphatase 2A (PP2A) activity. However, the effects of propofol, a common clinically used intravenous anesthetic, on tau phosphorylation under normothermic conditions are unknown. We investigated the effects of a general anesthetic dose of propofol on levels of phosphorylated tau in the mouse hippocampus and cortex under normothermic conditions. Thirty min following the administration of propofol 250 mg/kg i.p., significant increases in tau phosphorylation were observed at the AT8, CP13, and PHF-1 phosphoepitopes in the hippocampus, as well as at AT8, PHF-1, MC6, pS262, and pS422 epitopes in the cortex. However, we did not detect somatodendritic relocalization of tau. In both brain regions, tau hyperphosphorylation persisted at the AT8 epitope 2 h following propofol, although the sedative effects of the drug were no longer evident at this time point. By 6 h following propofol, levels of phosphorylated tau at AT8 returned to control levels. An initial decrease in the activity and expression of PP2A were observed, suggesting that PP2A inhibition is at least partly responsible for the hyperphosphorylation of tau at multiple sites following 30 min of propofol exposure. We also examined tau phosphorylation in SH-SY5Y cells transfected to overexpress human tau. A 1 h exposure to a clinically relevant concentration of propofol in vitro was also associated with tau hyperphosphorylation. These findings suggest that propofol increases tau phosphorylation both in vivo and in vitro under normothermic conditions, and further studies are warranted to determine the impact of this anesthetic on the acceleration of neurofibrillary pathology.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. Tau phosphorylation in mouse hippocampal…
Figure 1. Tau phosphorylation in mouse hippocampal tissue 30 min following the administration of propofol under hypothermic (A) or normothermic (B) conditions.
Hippocampal protein extracts were separated by SDS-PAGE and levels of tau phosphorylation were determined using antibodies directed at the AT8 (Ser202/Thr205;A1,B1), CP13 (Ser202;A2,B2), and PHF-1 (Ser396/Ser404;A3,B3) phosphoepitopes, or total tau (A4, B4). Relative immunoreactive band intensities are expressed as a percent of control (Ctl; intralipid) and are displayed for each phosphoepitope and total tau. For each condition, 2 representative data are displayed with Ctl (n = 4), and Prop (n = 5). Data are expressed as mean ± SD. *** denotes P<0.001, ** denotes P<0.01, and * denotes P<0.05 vs. Ctl with unpaired t-test.
Figure 2. Regional anatomical localization of tau…
Figure 2. Regional anatomical localization of tau phosphorylation following 30 min of anesthesia with propofol.
Fluorescence photomicrographs of hippocampal sagittal sections are shown with AT8 (Green, A,B,C), Total Tau (Red, D,E,F), or merged with DAPI (Green-Red-Blue, G,H,I), for the following conditions: Control (Intralipid, A,D,G), Hypothermia (B,E,H), and Normothermia (C,F,G). All images were taken at 5x magnification.
Figure 3. Tau phosphorylation in mouse hippocampal…
Figure 3. Tau phosphorylation in mouse hippocampal tissue 0.5 h and 2 h (A), or 0.5 h and 6 h (B) following the administration of propofol under normothermic conditions.
Hippocampal protein extracts were separated by SDS-PAGE and levels of tau phosphorylation were determined using antibodies directed at the AT8 (A1, B1), PHF-1 (A2, B2), and CP13 (A3, B3) phosphoepitopes, or Total Tau (A4, B4). Relative immunoreactive band intensities are expressed as a percent of control (Ctl; intralipid) and are displayed for each phosphoepitope and total tau. For each condition, 1 representative datum is displayed with Ctl (n = 4), 0.5 h (n = 5), 2 h (n = 5), or 6 h (n = 5). Data are expressed as mean ± SD. *, ** and *** denote P<0.05, P<0.01 and P<0.001 vs. ctl, respectively; ANOVA with Newman-Keuls post hoc test.
Figure 4. Tau phosphorylation in mouse cortical…
Figure 4. Tau phosphorylation in mouse cortical tissue 30 min and 2 h following the administration of propofol under normothermic conditions.
Cortical protein extracts were separated by SDS-PAGE and levels of tau phosphorylation were determined using antibodies directed at the AT8 (1), PHF-1 (2), pS262 (3), MC6 (4), and pS422 (5) phophoepitopes, or Total Tau (6). Relative immunoreactive band intensities are expressed as a percent of control (Ctl; intralipid) and are displayed for each phosphoepitope and total tau. For each condition, 1 representative datum is displayed with Ctl (n = 6), 0.5 h (n = 6), and 2 h (n = 7). Data are expressed as mean ± SD. *, and ** and denote P<0.05, and P<0.01 vs. Ctl, respectively; ANOVA with Newman-Keuls post hoc test.
Figure 5. Effect of propofol on tau…
Figure 5. Effect of propofol on tau kinases in mouse cortical tissue 0.5 h and 2 h following the administration of propofol under normothermic conditions.
Cortical protein extracts were separated by SDS-PAGE and levels of kinases were determined using antibodies directed at activated or total kinases as follow: (1) GSK-3β phospho-S9, (2) total GSK-3 (α and β), (3) phospho-ERK, (4) total ERK, (5) phospho-JNK, (6) total JNK, (7) phospho-CaMKII, (8) total CaMKII, (9) phospho-AKT, (10) total AKT, (11) CDK5, and (12) P35. Relative immunoreactive band intensities are expressed as a percent of control (Ctl; intralipid) and are displayed for each epitope. For each condition, 1 representative datum is displayed with Ctl (n = 6), 0.5 h (n = 6), and 2 h (n = 7). Data are expressed as mean ± SD. *, ** and *** denote P<0.05, P<0.01 and P<0.001 vs. ctl, respectively; ANOVA with Newman-Keuls post hoc test.
Figure 6. Effect of propofol on tau…
Figure 6. Effect of propofol on tau phosphatases in mouse cortical tissue 0.5 h and 2 h following the administration of propofol under normothermic conditions.
Cortical protein extracts were separated by SDS-PAGE and levels of phosphatases were determined using antibodies directed at the following proteins: (1) PP1 catalytic subunit, (2) PP2B catalytic subunit, and (3) PP2A catalytic subunit. Relative immunoreactive band intensities are expressed as a percent of control (Ctl; intralipid) and are displayed for each epitope. For each condition, 1 representative datum is displayed. (4) PP2A activity was measured with the PP2A Immunoprecipitation Phosphatase BioAssay Kit from US Biological and values expressed as percentage of control. All data are expressed as mean ± SD. * and ** denote P<0.05 and P<0.01 vs. ctl, respectively; Ctl (n = 6), 0.5 h (n = 6), and 2 h (n = 7); ANOVA with Newman-Keuls post hoc test.
Figure 7. Effect of propofol on tau…
Figure 7. Effect of propofol on tau phosphorylation in Tau-SH-SY5Y cells.
Cells were harvested after 1 h incubation at 37°C (n = 4) or 30°C (n = 3) in the absence of propofol (A), or following 1 h exposure to 10% intralipid in DMEM (Ctl, n = 3) or propofol (Prop, n = 5) at 3 µg/ml (16.8 µM), both at 37°C (B). Cell lysate protein extracts were separated by SDS-PAGE and the level of tau phosphorylation was determined using antibodies directed at the AT8 (A1, B1), CP13 (B2), or PHF-1 (B3) phosphoepitopes, or total tau (A2, B4). Relative immunoreactive band intensities are expressed as a percent of control and are displayed for each phosphoepitope and total tau. Tau phosphoepitopes are normalized on total tau. For each condition, 2 representative data are displayed. Data are expressed as mean ± SD, with *, **, and *** denoting P<0.05, P<0.01 and P<0.001 vs. ctl, respectively with unpaired t-test.

References

    1. Brookmeyer R, Johnson E, Ziegler-Graham K, Arrighi HM. Forecasting the global burden of Alzheimer's disease. Alzheimers Dement. 2007;3:186–191.
    1. Glenner GG, Wong CW. Alzheimer's disease: initial report of the purification and characterization of a novel cerebrovascular amyloid protein. Biochem Biophys Res Commun. 1984;120:885–890.
    1. Grundke-Iqbal I, Iqbal K, Quinlan M, Tung YC, Zaidi MS, et al. Microtubule-associated protein tau. A component of Alzheimer paired helical filaments. J Biol Chem. 1986;261:6084–6089.
    1. Grundke-Iqbal I, Iqbal K, Tung YC, Quinlan M, Wisniewski HM, et al. Abnormal phosphorylation of the microtubule-associated protein tau (tau) in Alzheimer cytoskeletal pathology. Proc Natl Acad Sci U S A. 1986;83:4913–4917.
    1. Montejo de Garcini E, Serrano L, Avila J. Self assembly of microtubule associated protein tau into filaments resembling those found in Alzheimer disease. Biochem Biophys Res Commun. 1986;141:790–796.
    1. Alonso A, Zaidi T, Novak M, Grundke-Iqbal I, Iqbal K. Hyperphosphorylation induces self-assembly of tau into tangles of paired helical filaments/straight filaments. Proc Natl Acad Sci U S A. 2001;98:6923–6928.
    1. Baranov D, Bickler PE, Crosby GJ, Culley DJ, Eckenhoff MF, et al. Consensus statement: First International Workshop on Anesthetics and Alzheimer's disease. Anesth Analg. 2009;108:1627–1630.
    1. Eckenhoff RG, Eckenhoff MF. Anesthesia, Amyloid and Alzheimer's. Cellscience Reviews. 2007:78–96.
    1. Run X, Liang Z, Gong CX. Anesthetics and Tau Protein: Animal Model Studies. J Alzheimers Dis. 2010.
    1. Papon MA, Whittington RA, El Khoury N, Planel E. Frontiers in Neurodegenerative Diseases in Press; 2011. Alzheimer's disease and anesthesia.
    1. Bianchi SL, Tran T, Liu C, Lin S, Li Y, et al. Brain and behavior changes in 12-month-old Tg2576 and nontransgenic mice exposed to anesthetics. Neurobiol Aging. 2008;29:1002–1010.
    1. Eckenhoff RG, Johansson JS, Wei H, Carnini A, Kang B, et al. Inhaled anesthetic enhancement of amyloid-beta oligomerization and cytotoxicity. Anesthesiology. 2004;101:703–709.
    1. Xie Z, Dong Y, Maeda U, Alfille P, Culley DJ, et al. The common inhalation anesthetic isoflurane induces apoptosis and increases amyloid beta protein levels. Anesthesiology. 2006;104:988–994.
    1. Xie Z, Dong Y, Maeda U, Moir RD, Xia W, et al. The inhalation anesthetic isoflurane induces a vicious cycle of apoptosis and amyloid beta-protein accumulation. J Neurosci. 2007;27:1247–1254.
    1. Planel E, Richter KE, Nolan CE, Finley JE, Liu L, et al. Anesthesia leads to tau hyperphosphorylation through inhibition of phosphatase activity by hypothermia. J Neurosci. 2007;27:3090–3097.
    1. Planel E, Krishnamurthy P, Miyasaka T, Liu L, Herman M, et al. Anesthesia-induced hyperphosphorylation detaches 3-repeat tau from microtubules without affecting their stability in vivo. J Neurosci. 2008;28:12798–12807.
    1. Planel E, Bretteville A, Liu L, Virag L, Du AL, et al. Acceleration and persistence of neurofibrillary pathology in a mouse model of tauopathy following anesthesia. Faseb J. 2009 doi: .
    1. Run X, Liang Z, Zhang L, Iqbal K, Grundke-Iqbal I, et al. Anesthesia induces phosphorylation of tau. J Alzheimers Dis. 2009;16:619–626.
    1. Wunsch H, Kahn JM, Kramer AA, Rubenfeld GD. Use of intravenous infusion sedation among mechanically ventilated patients in the United States. Crit Care Med. 2009;37:3031–3039.
    1. Su JH, Cummings BJ, Cotman CW. Early phosphorylation of tau in Alzheimer's disease occurs at Ser-202 and is preferentially located within neurites. Neuroreport. 1994;5:2358–2362.
    1. Zhu X, Lee HG, Raina AK, Perry G, Smith MA. The role of mitogen-activated protein kinase pathways in Alzheimer's disease. Neurosignals. 2002;11:270–281.
    1. Planel E, Sun X, Takashima A. Role of GSK-3 beta in Alzheimer's disease pathology. Drug Development Research. 2002;56:491–510.
    1. Maccioni RB, Otth C, Concha, Munoz JP. The protein kinase Cdk5. Structural aspects, roles in neurogenesis and involvement in Alzheimer's pathology. Eur J Biochem. 2001;268:1518–1527.
    1. Sadik G, Tanaka T, Kato K, Yamamori H, Nessa BN, et al. Phosphorylation of tau at Ser214 mediates its interaction with 14-3-3 protein: implications for the mechanism of tau aggregation. J Neurochem. 2009;108:33–43.
    1. Cross DA, Alessi DR, Cohen P, Andjelkovich M, Hemmings BA. Inhibition of glycogen synthase kinase-3 by insulin mediated by protein kinase B. Nature. 1995;378:785–789.
    1. Litersky JM, Johnson GV, Jakes R, Goedert M, Lee M, et al. Tau protein is phosphorylated by cyclic AMP-dependent protein kinase and calcium/calmodulin-dependent protein kinase II within its microtubule-binding domains at Ser-262 and Ser-356. Biochem J. 1996;316:655–660.
    1. Wang JZ, Grundke-Iqbal I, Iqbal K. Restoration of biological activity of Alzheimer abnormally phosphorylated tau by dephosphorylation with protein phosphatase-2A, - 2B and -1. Brain Res. 1996;38:200–208.
    1. Goedert M, Cohen ES, Jakes R, Cohen P. p42 MAP kinase phosphorylation sites in microtubule-associated protein tau are dephosphorylated by protein phosphatase 2A1. Implications for Alzheimer's disease [corrected] [published erratum appears in FEBS Lett 1992 Nov 23;313(2):203]. FEBS Lett. 1992;312:95–99.
    1. Engdahl O, Abrahams M, Bjornsson A, Vegfors M, Norlander B, et al. Cerebrospinal fluid concentrations of propofol during anaesthesia in humans. Br J Anaesth. 1998;81:957–959.
    1. Planel E, Miyasaka T, Launey T, Chui DH, Tanemura K, et al. Alterations in glucose metabolism induce hypothermia leading to tau hyperphosphorylation through differential inhibition of kinase and phosphatase activities: implications for Alzheimer's disease. J Neurosci. 2004;24:2401–2411.
    1. Pei JJ, Gong CX, An WL, Winblad B, Cowburn RF, et al. Okadaic-acid-induced inhibition of protein phosphatase 2A produces activation of mitogen-activated protein kinases ERK1/2, MEK1/2, and p70 S6, similar to that in Alzheimer's disease. Am J Pathol. 2003;163:845–858.
    1. Singh TJ, Grundke-Iqbal I, McDonald B, Iqbal K. Comparison of the phosphorylation of microtubule-associated protein tau by non-proline dependent protein kinases. Mol Cell Biochem. 1994;131:181–189.
    1. Tian Q, Wang J. Role of serine/threonine protein phosphatase in Alzheimer's disease. Neurosignals. 2002;11:262–269.
    1. Bennecib M, Gong CX, Grundke-Iqbal I, Iqbal K. Role of protein phosphatase-2A and -1 in the regulation of GSK-3, cdk5 and cdc2 and the phosphorylation of tau in rat forebrain. FEBS Lett. 2000;485:87–93.
    1. Gong CX, Lidsky T, Wegiel J, Zuck L, Grundke-Iqbal I, et al. Phosphorylation of microtubule-associated protein tau is regulated by protein phosphatase 2A in mammalian brain. Implications for neurofibrillary degeneration in Alzheimer's disease. J Biol Chem. 2000;275:5535–5544.
    1. Planel E, Yasutake K, Fujita SC, Ishiguro K. Inhibition of protein phosphatase 2A overrides tau protein kinase I/glycogen synthase kinase 3 beta and cyclin-dependent kinase 5 inhibition and results in tau hyperphosphorylation in the hippocampus of starved mouse. J Biol Chem. 2001;276:34298–34306.
    1. Ehlenbach WJ, Hough CL, Crane PK, Haneuse SJ, Carson SS, et al. Association between acute care and critical illness hospitalization and cognitive function in older adults. JAMA. 2010;303:763–770.
    1. Smith PK, Krohn RI, Hermanson GT, Mallia AK, Gartner FH, et al. Measurement of protein using bicinchoninic acid. Anal Biochem. 1985;150:76–85.
    1. Delobel P, Flament S, Hamdane M, Mailliot C, Sambo AV, et al. Abnormal Tau phosphorylation of the Alzheimer-type also occurs during mitosis. J Neurochem. 2002;83:412–420.
    1. Hamdane M, Sambo AV, Delobel P, Begard S, Violleau A, et al. Mitotic-like tau phosphorylation by p25-Cdk5 kinase complex. J Biol Chem. 2003;278:34026–34034.
    1. Mailliot C, Bussiere T, Hamdane M, Sergeant N, Caillet ML, et al. Pathological tau phenotypes. The weight of mutations, polymorphisms, and differential neuronal vulnerabilities. Ann N Y Acad Sci. 2000;920:107–114.
    1. Oscarsson A, Juhas M, Sjolander A, Eintrei C. The effect of propofol on actin, ERK-1/2 and GABAA receptor content in neurones. Acta Anaesthesiol Scand. 2007;51:1184–1189.
    1. Goedert M, Jakes R, Vanmechelen E. Monoclonal antibody AT8 recognises tau protein phosphorylated at both serine 202 and threonine 205. Neurosci Lett. 1995;189:167–169.
    1. Otvos L, Jr, Feiner L, Lang E, Szendrei GI, Goedert M, et al. Monoclonal antibody PHF-1 recognizes tau protein phosphorylated at serine residues 396 and 404. J Neurosci Res. 1994;39:669–673.
    1. Jicha GA, Lane E, Vincent I, Otvos L, Jr, Hoffmann R, et al. A conformation- and phosphorylation-dependent antibody recognizing the paired helical filaments of Alzheimer's disease. J Neurochem. 1997;69:2087–2095.
    1. Weaver CL, Espinoza M, Kress Y, Davies P. Conformational change as one of the earliest alterations of tau in Alzheimer's disease. Neurobiol Aging. 2000;21:719–727.
    1. Jicha GA, Bowser R, Kazam IG, Davies P. Alz-50 and MC-1, a new monoclonal antibody raised to paired helical filaments, recognize conformational epitopes on recombinant tau. J Neurosci Res. 1997;48:128–132.
    1. Planel E, Tatebayashi Y, Miyasaka T, Liu L, Wang L, et al. Insulin dysfunction induces in vivo tau hyperphosphorylation through distinct mechanisms. J Neurosci. 2007;27:13635–13648.

Source: PubMed

3
Tilaa