Next Article in Journal / Special Issue
Interactions between Endostatin and Vascular Endothelial Growth Factor (VEGF) and Inhibition of Choroidal Neovascularization
Previous Article in Journal / Special Issue
Electrochemical Studies of Camptothecin and Its Interaction with Human Serum Albumin
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Hsp90 Maintains the Stability and Function of the Tau Phosphorylating Kinase GSK3β

Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, Southeast University Medical School, Nanjing 210009, P.R.China
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2007, 8(1), 51-60; https://doi.org/10.3390/i8010060
Submission received: 2 January 2007 / Accepted: 23 January 2007 / Published: 30 January 2007
(This article belongs to the Special Issue Interaction of Biological Molecules)

Abstract

:
Hyperphosphorylation of tau leading to aggregated tau and tangle formation is a common pathological feature of tauopathies, including Alzheimer’s disease. Abnormal phosphorylation of tau by kinases, in particular GSK3β, has been proposed as a pathogenic mechanism in these diseases. In this study we demonstrate that the heat shock protein 90 (Hsp90) maintains the stability and function of the GSK3β. By using both rat primary cortical neurons and COS-7 cells, we show that Hsp90 inhibitors lead to a reduction of the protein level of GSK3β, and that this effect is associated with both a decrease in tau phosphorylation at putative GSK3β sites and an induction in heat shock protein 70 (Hsp70) levels. We further show that Hsp90 associates with the GSK3β regulating its stability and function and preventing its degradation by the proteasome.

1. Introduction

The heat shock proteins (Hsps) are a family of ubiquitous and abundant proteins found in all eukaryotes and prokaryotes. Their function is to mediate the proper folding of native and denatured proteins to insure that those proteins maintain their native conformation [1, 2]. In addition to their functions in co- and posttranslational folding, Hsps are also required for intracellular protein trafficking and translocation, are involved in macromolecular assembly and disassembly, and facilitate the protein degradation through the ubiquitin-proteasome pathway [3, 4]. All those functions of Hsps are well characterized and are central to maintaining cellular homeostasis and in promoting cell survival in response to stressful conditions.
During about a century of the research on neurodegenerative diseases, researchers find that a common histological feature shared by those diseases is the existence of intra-cellular and/or extra-cellular aggregations in the brain. The appearance of aggregates in diseased brain implies an underlying incapacity of molecular chaperones [57]. Based on this idea, the relationship between molecular chaperones, particularly heat shock proteins, and neurodegenerative disorders has been thoroughly studied. It has been demonstrated that Hsps could suppress neurodegeneration in cultured cell, fly and mouse models of Huntington’s disease, Parkingson’s disease and Alzheimer’s disease [813]. While it is becoming clear that Hsps can have a profound influence on solubility and aggregation of proteins that involved in various neurodegenerative diseases, many other important roles Hsps may play remain to be examined. After all, Hsps are a group of multi-function proteins. For instance, the involvement of Hsp90 in many signal transduction pathways has been well established, Hsp90 directly associates with several important protein kinases such as Akt, ErbB2, Polo kinase and Raf1 [1417]. Hsp90 inhibitors could induce the inactivation/degradation of those client proteins. In our previous study we have reported that inducing the expression of Hsps can reduce tau phosphorylation [13], and a subsequent study showed that Hsp90 inhibitors could cause the inactivation of ERK by facilitating the degradation of Raf [18]. In this study we report that Hsp90 could also regulate the cellular stability of GSK3β, an important protein kinase account for tau hyperphosphorylation. Our results show that treatment of cells with Hsp90 inhibitors results in the proteasome-dependent degradation of GSK3β, and subsequently cause the reduction of tau phosphorylation.

2. Materials and Methods

Geldanamycin (GA), ZVAD-FMK, pepstatin, MG132, leupeptin, E64 and Protein A/G plus agarose beads were purchased from Calbiochem, San Diego, CA; poly-L-lysine, novobicin are ordered from Sigma; BCA protein assay kit is ordered from Pierce; FuGENE 6 reagent is ordered from Roche Molecular Biochemicals; cDNA constructs expressing human wild type Tau (T40) and was a kind gift from Dr. Virginia Lee (University of Pennsylvania, Philadelphia, PA); PU24FCl was a kind gift from Dr. Gabriela Chiosis (Memorial Sloan-Kettering Cancer Center, New York, NY); Dulbecco’s Modified Eagle Medium (DMEM), neurobasal medium, fetal bovine serum (FBS), penicillin/streptomycin and B27 supplement are ordered from Invitrogen; anti-β-actin mouse monoclonal antibody (A1978) is ordered from Sigma, St. Louis, MO; anti-GSK3β (#9332) antibody is ordered from cell signaling; antiphospho-tau (pS199/202) is ordered from Biosource; AT8, AT180, AT100, AT270 are ordered from Innogenetics; anti-tau abtibody is from Santa Cruz; anti-Hsp70 (SPA-810) and anti-Hsp90 (SPA-830) are from Stressgen Bioreagents, Victoria, British Columbia, Canada.

2.1 COS-7 Cell Cultures and Incubation with the Hsp90 Inhibitor

COS-7 cells grown in DMEM with 10% FBS and penicillin/streptomycin (50 U/50μg/ml, respectively) were transiently transfected using FuGENE 6 reagent to over-express wild type hTau. At 12 h after transfection, cells were incubated for 20h with the indicated concentration of GA or a purine-based Hsp90 inhibitor PU24FCl [19]. Cells were harvested and lysed in 1 × RIPA buffer (50mM Tris-HCl, 150 mM NaCl, 1% Nonidet P-40, 5 mM EDTA, 0.5% sodium deoxycholate, 0.1% SDS, pH 8.0). The protein concentration was determined using BCA assay. Samples with equal amounts of protein were analyzed by Western blotting.

2.2 Primary Neuronal Cultures and Incubation with the Hsp90 Inhibitor

Primary neuronal cultures were derived from the cerebral cortices of embryonic day 17 (E17) rat embryos and maintained as previously described [20]. Dissociated neurons were plated (~5 × 106 cells per plate) on poly-L-lysine pre-coated 6cm plates in serum-free neurobasal medium with N2 and B27 supplements and cultured for 7 days. Media were replaced every 2 days with the addition of 16.5 mg/ml uridine and 6.7 mg/ml 5-fluoro-2′-deoxyuridine to prevent proliferation of glial cells. Hsp90 inhibitor was added at day 6 of culture and cells were incubated at 37 °C as indicated. Following incubation, cells were collected and lysed. Samples were analyzed by SDS/PAGE and Western blotting. For studies involving inhibitors of protein degradation, primary cortical cultures were treated with DMSO or indicated concentration of Hsp90 inhibitor in the presence of the following protease inhibitors: 10 μM MG132, 1μM pepstatin, 75μM leupeptin, 10 μM E64 or 20 μM ZVAD for 20h. Cells were collected, and the lysates analyzed by Western blotting.

2.3 Cycloheximide Treatments

Primary cortical cultures or COS-7 transfected cells were treated with cycloheximide (at a final concentration of 100 μg/ml) for the indicated times. Cell lysates were run on SDS-PAGE gel and proteins transferred to PVDF membranes to be probed with anti-GSK3β antibody.

2.4 Immunoprecipitation

For isolation of the GSK3β/Hsp90 complexes from cultured cells, COS-7 cells were lysed in TMNSV buffer (50 mM Tris-HCl, pH7.0, 20 mM Na2MoO4, 0.09% NP-40, 150 mM NaCl, 1 mM sodium othovanadate with added protease inhibitors mixture). Cell nuclei and debris were removed by centrifuging at 13,000 × g for 5 min. Samples were immunoprecipitated with anti-GSK3β antibody for 4h at 4°C. Immunoreactive materials and protein A/G agarose beads complexes were washed three times with lysis buffer and once with PBS. Immunoreactive materials were eluted from the beads by incubating with sample buffer for 5min at 95°C and subjected to SDS/PAGE followed by Western blotting.

3. Results

3.1 Inhibition of Hsp90 reduces tau phosphorylation level and protein level of GSK3β

COS cells transfected with wild type hTau (the longest isform 1-441aa) were treated with two different Hsp90 inhibitors, GA and PU24FCl. Both inhibitors bind selectively to the regulatory pocket of Hsp90 which inhibits chaperone function and alter the association of client proteins with the chaperone complex. As a consequence, these proteins do not achieve their mature functional conformation and are degraded by the proteasome. In our previous paper, we reported that the inhibition of Hsp90 resulted in a reduction of the protein level of mutant tau. While in the case of WT tau protein, the inhibition of Hsp90 has no effect on total protein level, but to decrease the tau phosphorylation instead (Figure 1, Figure 3). Tau protein could be phosphorylated by several kinases at dozens sites, but only a few kinases have been confirmed related to the pathogenesis of AD, GSK3β is one of such kinases [21]. Several sites which phosporylation level could be regulated by Hsp90 inhibitor in this study are putative GSK3β sites [22]. In order to study whether the reduction of tau phosphorylation is due to the inactive/degradation of GSK3β, the total GSK3β protein level has been checked in COS cells and rat primary cortical neurons. After treated with 10μM PU24FCl or 400nM GA for 20h, the total GSK3β level in both COS cell and primary neurons are decreased dramatically (Figure 2). At the same time, the Hsp70 expression was induced because the heat shock factor 1 (HSF1) is released from Hsp90 complex as documented by Zou et al [23]. By using another Hsp90 inhibitor, novobicin, which binds to Hsp90 at c-terminus rather than the regular ATP pocket, we also observed a dose –dependent reduction of GSK3β (Figure 2).
The effect of Hsp90 inhibitors on kinases was well-defined and selective as the expression of casein kinase 1 (CK-1), casein kinase 2 (CK-2) and cyclic AMP-dependent protein kinase (PKA), kinases shown to prime Tau for GSK3β phosphorylation [24] was not affected by the Hsp90 inhibitor (data not shown).

3.2 Degradation of GSK3β by Hsp90 inhibition is mediated by the proteasome

Protein degradation may be achieved in cells by several protease systems. However, proteins regulated by Hsp90 appear to be degraded majorly via the proteasome pathway upon chaperone inhibition [25]. To examine whether the proteasome is accounted for the GSK3β degradation in our study, we explored the effect of several inhibitors of other proteases in preventing the degradation of GSK3 by Hsp90 inhibitors. Inhibitors of lysosome proteases (E64 and leupeptin), aspartate proteases (pepstatin) or caspases (ZVAD) failed to rescue GSK3β. Only a proteasome inhibitor (MG132) was able to prevent their degradation over a 20 h treatment with the Hsp90 inhibitor (Figure 3). These observations are in agreement with observed effects on other oncoprotein clients of Hsp90, which suggest that GSK3β are protected through Hsp90 chaperone complex formation, from ubiquitination and proteasomal degradation. In concordance with this result, the decrease of tau phosphorylation could not be prevented by those protease inhibitors as well except MG132 (Figure 3). The effect of MG132 on tau phosphorylation is complicate because the MG132 itself could cause the tau phosphorylation decrease [26]. Under this circumstance it’s hard to distinguish the effect of Hsp90 inhibitor from proteolytic stress. Maybe both pathways share the same mechanism to reduce tau phosphorylation, since MG132 treatment could induce heat shock response in cells as well [26]. A rational hypothesis is that proteolytic stress results in a increase of misfolded proteins in cytosol, which competed with Hsp90 client proteins for this chaperone. Those client proteins of Hsp90 include HSF1 and GSK3, may head to different destination. HSF1 moves to nucleus and activates the expression of Hsps, while the GSK3 and other kinases head to ubiquitination.

3.3 Hsp90 regulates the stability of GSK3β

To examine if Hsp90 plays a direct role in maintaining the stability of GSK3β, we tested whether inhibition of Hsp90 function affected its half-life. Primary neuronal cultures were treated with inhibitor or vehicle in the presence of cycloheximide, a protein synthesis inhibitor. The half-life of the GSK3β protein was remarkably shortened in the cells treated with Hsp90 inhibitors (Figure 4). Moreover, the protein was degraded upon the Hsp90 inhibitor treatment even when induction of Hsp70 was blocked by the protein-synthesis inhibitor. These findings strongly implicate Hsp90 as a direct and important regulator of GSK3β stability.

3. 4 GSK3β exist in a complex with Hsp90

We next immunoprecipitated endogenous GSK3β from 2mg of COS cells lysate and demonstrated by western blotting that Hsp90 was coimmunoprecipitated. During the experiment, 15μg of COS cells lysate was loading on the same gel and immunoblotted in parallel. The result showed that only a small fraction of cellular Hsp90 is associated with GSK3β. This observation is not surprising as the Hsp90 is one of the most abundant cellular proteins and is known to be associated with a large number of proteins.

Discussion

Hyperphosphorylation of tau leading to aggregated tau and tangle formation is a common pathological feature of tauopathies, including Alzheimer’s disease. The toxic behavior of the abnormal tau is solely due to its hyperphosphorylation because dephosphorylation restores it into a normal-like protein [27]. The longest form of human brain tau contains 80 serine and threonine and 5 tyrosine residues. Thus, almost 20% of the protein has the potential to become phosphorylated. Several kinases, including cyclin-dependent kinase 5 (Cdk5) and glycogen synthase kinase-3 beta (GSK3β) have been implicated in phosphorylating tau at pathogenic sites. Abnormal phosphorylation of tau by kinases, in particular the GSK3β and Cdk5, has been proposed as a pathogenic mechanism in these diseases [2830]. Cells co-transfected with GSK3β and tau showed abnormal microtubule bundles [31]. Recent animal studies have provided further support for the view that GSK3β is the most likely candidate for the protein kinase responsible for the abnormal phosphorylation state of tau in AD brain. Immunohistochemical studies located GSK3 in neurofibrillary tangles [32]. Moreover, GSK3β is elevated in AD human brain [33]. Maintenance of such anomalously activated kinases in a functional state requires a stabilizing mechanism. In oncogenic transformation, a corresponding role is assigned to molecular chaperones, in particular to the heat shock protein 90 (Hsp90) [3436]. This chaperone allows cancer cells to tolerate the many components of dysregulated pathways in a transformation-specific manner by interacting with several client substrates, such as kinases, hormone receptors and transcription factors that are directly involved in driving multistep malignancy, and also with mutated oncogenic proteins required for the transformed phenotype. Under normal conditions, Hsp90 interacts with these client proteins in a dynamic, low-affinity manner. Upon mutation or dysregulated function many of these client proteins display unusually stable associations with Hsp90-containing chaperone complexes. These associations maintain them capable to function in the dysregulated state and appear to be essential for their transforming, aberrant activity. A similar role for Hsp90 in controlling kinases may be envisioned in neurodegenerative diseases. In this study we demonstrate that the heat shock protein 90 (Hsp90) maintains the stability and function of the GSK3β. By using both rat primary cortical neurons and COS-7 cells, we show that Hsp90 inhibitors lead to a destabilization of GSK3β, and that this effect is associated with both a decrease in tau phosphorylation at putative GSK3β. We further show that Hsp90 associates with the GSK3β regulating its stability and function and preventing its degradation by the proteasome.
Direct inhibition of kinases as a therapeutic modality in tauopathies has been recently investigated. Inhibition of GSK3β by lithium was demonstrated to reduced tauopathy and degeneration in the P301L tau transgenic mouse model [37]. These results support the idea that kinases are involved in tauopathy progression and that kinase inhibitors may be effective therapeutically. We propose here indirect kinase inhibition by interference with Hsp90 activity as another therapeutic strategy. Considering the facts that Hsp90 inhibitors have entered the phase I/II clinical trial for cancer, our work opens the door to the exciting possibility of using these emerging agents in the treatment of neurodegenerative diseases.

Acknowledgements

We thank Dr. V. Lee for the Tau constructs, Dr. G. Chiosis for the PU24FCl compound. This work was supported by National Natural Science Foundation of China Grant 30370311(to F. D.).

References and Notes

  1. Hartl, F.U.; Hayer-Hartl, M. Molecular chaperones in the cytosol: from nascent chain to folded protein. Science 2002, 295(5561), 1852–1858. [Google Scholar]
  2. Young, J.C.; et al. Pathways of chaperone-mediated protein folding in the cytosol. Nat Rev Mol Cell Biol 2004, 5(10), 781–791. [Google Scholar]
  3. Goldberg, A.L. Protein degradation and protection against misfolded or damaged proteins. Nature 2003, 426(6968), 895–899. [Google Scholar]
  4. Muchowski, P.J. Protein misfolding, amyloid formation, and neurodegeneration: a critical role for molecular chaperones? Neuron 2002, 35(1), 9–12. [Google Scholar]
  5. Sherman, M.Y.; Goldberg, A.L. Cellular defenses against unfolded proteins: a cell biologist thinks about neurodegenerative diseases. Neuron 2001, 29(1), 15–32. [Google Scholar]
  6. Muchowski, P.J.; Wacker, J.L. Modulation of neurodegeneration by molecular chaperones. Nat Rev Neurosci 2005, 6(1), 11–22. [Google Scholar]
  7. Slavotinek, AM; Biesecker, LG. Unfolding the role of chaperones and chaperonins in human disease. Trends Genet 2001, 17(9), 528–535. [Google Scholar]
  8. Carmichael, J; Chatellier, J; Woolfson, A; Milstein, C; Fersht, AR; Rubinsztein, DC. Bacterial and yeast chaperones reduce both aggregate formation and cell death in mammalian cell models of Huntington’s disease. Proc. Natl. Acad. Sci. USA 2000, 97(17), 9701–9705. [Google Scholar]
  9. Warrick, JM; Chan, HY; Gray-Board, GL; Chai, Y; Paulson, HL; Bonini, NM. Suppression of polyglutamine-mediated neurodegeneration in Drosophila by the molecular chaperone HSP70. Nat. Genet 1999, 23(4), 425–428. [Google Scholar]
  10. Sittler, A; Lurz, R; Lueder, G; Priller, J; Lehrach, H; Hayer-Hartl, MK; Hartl, FU; Wanker, EE. Geldanamycin activates a heat shock response and inhibits huntingtin aggregation in a cell culture model of Huntington’s disease. Hum. Mol. Genet 2001, 10(16), 1307–1315. [Google Scholar]
  11. Auluck, PK; Chan, HY; Trojanowski, JQ; Lee, VM; Bonini, NM. Chaperone suppression of alpha-synuclein toxicity in a Drosophila model for Parkinson’s disease. Science 2002, 295(5556), 865–868. [Google Scholar]
  12. Muchowski, PJ; Schaffar, G; Sittler, A; Wanker, EE; Hayer-Hartl, MK; Hartl, FU. Hsp70 and hsp40 chaperones can inhibit self-assembly of polyglutamine proteins into amyloid-like fibrils. Proc. Natl. Acad. Sci. USA 2000, 97(14), 7841–7846. [Google Scholar]
  13. Dou, F; Netzer, WJ; Tanemura, K; Li, F; Hartl, FU; Takashima, A; Gouras, GK; et al. Chaperones increase association of tau protein with microtubules. Proc. Natl. Acad. Sci. USA 2003, 100(2), 721–726. [Google Scholar]
  14. Xu, W; Mimnaugh, E; Rosser, MF; Nicchitta, C; Marcu, M; Yarden, Y; Neckers, L. Sensitivity of mature Erbb2 to geldanamycin is conferred by its kinase domain and is mediated by the chaperone protein Hsp90. J. Biol. Chem 2001, 276(5), 3702–3708. [Google Scholar]
  15. Sato, S; Fujita, N; Tsuruo, T. Modulation of Akt kinase activity by binding to Hsp90. Proc. Natl Acad. Sci. USA 2000, 97(20), 10832–10837. [Google Scholar]
  16. Schulte, TW; Blagosklonny, MV; Ingui, C; Neckers, L. Disruption of the Raf-1-Hsp90 molecular complex results in destabilization of Raf-1 and loss of Raf-1-Ras association. J. Biol. Chem 1995, 270(41), 24585–24588. [Google Scholar]
  17. de Carcer, G; do Carmo Avides, M; Lallena, MJ; Glover, DM; Gonzalez, C. Requirement of Hsp90 for centrosomal function reflects its regulation of Polo kinase stability. Eur. Mol. Biol. Org. J 2001, 20(11), 2878–2884. [Google Scholar]
  18. Dou, F.; Yuan, L.D.; Zhu, J.J. Heat shock protein 90 indirectly regulates ERK activity by affecting Raf protein metabolism. Acta Biochim Biophys Sin (Shanghai) 2005, 37(7), 501–505. [Google Scholar]
  19. Vilenchik, M.; et al. Targeting wide-range oncogenic transformation via PU24FCl, a specific inhibitor of tumor Hsp90. Chem. Biol 2004, 11, 787–797. [Google Scholar]
  20. Xu, H.; et al. Estrogen reduces neuronal generation of Alzheimer beta-amyloid peptides. Nat Med 1998, 4(4), 447–51. [Google Scholar]
  21. Lee, V.M.; Goedert, M.; Trojanowski, J.Q. Neurodegenerative tauopathies, in. Annu Rev Neurosci 2001, 1121–1159. [Google Scholar]
  22. Billingsley, M.L.; Kincaid, R.L. Regulated phosphorylation and dephosphorylation of tau protein: effects on microtubule interaction, intracellular trafficking and neurodegeneration. Biochem J 1997, 323 Pt 3, 577–91. [Google Scholar]
  23. Zou, J; Guo, Y; Guettouche, T; Smith, DF; Voellmy, R. Repression of heat shock transcription factor HSF1 activation by HSP90 (HSP90 complex) that forms a stress-sensitive complex with HSF1. Cell 1998, 94(4), 471–480. [Google Scholar]
  24. Singh, T. J.; Zaidi, T.; Grundke-Iqbal, I.; Iqbal, K. Modulation of GSK-3-catalyzed phosphorylation of microtubule-associated protein tau by non-prolinedependent protein kinases. FEBS Lett 1995, 358, 4–8. [Google Scholar]
  25. Mimnaugh, E.; Chavany, C.; Neckers, L. M. Polyubiquitination and proteasomal degradation of the p185c-erbB-2 receptor protein-tyrosine kinase induced by geldanamycin. J. Biol. Chem 1996, 271, 22796–22801. [Google Scholar]
  26. Goldbaum, O; Richter-Landsberg, C. Proteolytic stress causes heat shock protein induction, tau ubiquitination, and the recruitment of ubiquitin to tau-positive aggregates in oligodendrocytes in culture. J Neurosci 2004, 24(25), 5748–5757. [Google Scholar]
  27. Iqbal, K; Grundke-Iqbal, I. Inhibition of neurofibrillary degeneration: a promising approach to Alzheimer’s disease and other tauopathies. Curr Drug Targets 2004, 5(6), 495–502. [Google Scholar]
  28. Stoothoff, WH; Johnson, GV. Tau phosphorylation: physiological and pathological consequences. Biochim Biophys Acta 2005, 1739(2–3), 280–297. [Google Scholar]
  29. Monaco, EA, 3rd; Vallano, ML. Role of protein kinases in neurodegenerative disease: cyclin-dependent kinases in Alzheimer’s disease. Front Biosci 2005, 10, 143–159. [Google Scholar]
  30. Kosik, K. S.; Shimura, H. Phosphorylated tau and the neurodegenerative foldopathies. Biochim. Biophys. Acta 2005, 1739, 298–310. [Google Scholar]
  31. Sang, H.; et al. Phosphorylation of tau by glycogen synthase kinase 3β in intact mammalian cells influences the stability of microtubules. Neurosci. Lett 2001, 312, 141–144. [Google Scholar]
  32. Pei, J.J.; et al. Distribution of active glycogen synthase kinase 3β (GSK-3β) in brains staged for Alzheimer disease neurofibrillary changes. J. Neuropathol. Exp. Neurol 1999, 58, 1010–1019. [Google Scholar]
  33. Yamaguchi, H.; et al. Preferential labeling of Alzheimer neurofibrillary tangles with antisera for tau protein kinase (TPK) I/glycogen synthase kinase-3 β and cyclin-dependent kinase 5, a component of TPK II. Acta Neuropathol 1996, 92, 232–241. [Google Scholar]
  34. Neckers, L. Chaperoning oncogenes: Hsp90 as a target of geldanamycin. Handb. Exp. Pharmacol 2006, 172, 259–77. [Google Scholar]
  35. McDonald, E.; Workman, P.; Jones, K. Inhibitors of the HSP90 molecular chaperone: attacking the master regulator in cancer. Curr. Top. Med. Chem 2006, 6, 1091–1107. [Google Scholar]
  36. Chiosis, G. Targeting chaperones in transformed systems--a focus on Hsp90 and cancer. Expert Opin. Ther. Targets 2006, 10, 37–50. [Google Scholar]
  37. Noble, W; Planel, E; Zehr, C; Olm, V; Meyerson, J; Suleman, F; Gaynor, K; Wang, L; LaFrancois, J; Feinstein, B; Burns, M; Krishnamurthy, P; Wen, Y; Bhat, R; Lewis, J; Dickson, D; Duff, K. Inhibition of glycogen synthase kinase-3 by lithium correlates with reduced tauopathy and degeneration in vivo. Proc Natl Acad Sci U S A 2005, 102(19), 6990–6995. [Google Scholar]
Figure 1. Effects of Hsp90 inhibition on tau phosphorylation. COS-7 cells were transiently transfected to over-express human tau protein longest isoform. At 12 h after transfection, cells were incubated for 20h with10μM PU24FCl or 400nM GA, cells were collected and lysed, the resulting lysates analyzed by immunoblot.
Figure 1. Effects of Hsp90 inhibition on tau phosphorylation. COS-7 cells were transiently transfected to over-express human tau protein longest isoform. At 12 h after transfection, cells were incubated for 20h with10μM PU24FCl or 400nM GA, cells were collected and lysed, the resulting lysates analyzed by immunoblot.
Ijms 08 00051f1
Figure 2. Inhibition of Hsp90 specifically decreases GSK3β. COS cells were treated with10μM PU24FCl or 400nM GA for 20h, in some experiments, cells were treated with novobicin at indicated concentration for 20h. Rat embryonic cortical primary neurons were treated under same conditions.
Figure 2. Inhibition of Hsp90 specifically decreases GSK3β. COS cells were treated with10μM PU24FCl or 400nM GA for 20h, in some experiments, cells were treated with novobicin at indicated concentration for 20h. Rat embryonic cortical primary neurons were treated under same conditions.
Ijms 08 00051f2
Figure 3. Degradation of GSK3β by Hsp90 inhibitors is mediated by the proteasome. Only proteasome inhibitors (i.e. MG132) were able to prevent the degradation of GSK3β over a 20 h treatment with the Hsp90 inhibitors PU24FCl and GA. Primary neurons were treated with DMSO, 10 μM PU24FCl or 400nM GA in the presence of the following inhibitors: 10 μM MG132, 1μM pepstatin, 75μM leupeptin, 10 μM E64 or 20 μM ZVAD for 20h. Cells were collected, lysed and the resulting lysates analyzed by immunoblot. Proteins were analyzed by Western blot. The phosporylation level of tau protein was analyzed by Western blotting using phospho-specific antibodies [AT8 (1:250), AT100 (1:250), AT180(1:250), AT270 (1:250)] anti total tau antibody (1:500). Actin were used as protein quantification control.
Figure 3. Degradation of GSK3β by Hsp90 inhibitors is mediated by the proteasome. Only proteasome inhibitors (i.e. MG132) were able to prevent the degradation of GSK3β over a 20 h treatment with the Hsp90 inhibitors PU24FCl and GA. Primary neurons were treated with DMSO, 10 μM PU24FCl or 400nM GA in the presence of the following inhibitors: 10 μM MG132, 1μM pepstatin, 75μM leupeptin, 10 μM E64 or 20 μM ZVAD for 20h. Cells were collected, lysed and the resulting lysates analyzed by immunoblot. Proteins were analyzed by Western blot. The phosporylation level of tau protein was analyzed by Western blotting using phospho-specific antibodies [AT8 (1:250), AT100 (1:250), AT180(1:250), AT270 (1:250)] anti total tau antibody (1:500). Actin were used as protein quantification control.
Ijms 08 00051f3
Figure 4. Hsp90 inhibition affects the protein’s half-life. COS-7 cells were treated with DMSO or 10 μM PU24FCl in the presence of 30 μM cyclohexamide to halt translation. At the times indicated, cells were lysed and whole-cell lysates immunobloted with anti-GSK3β antibody.
Figure 4. Hsp90 inhibition affects the protein’s half-life. COS-7 cells were treated with DMSO or 10 μM PU24FCl in the presence of 30 μM cyclohexamide to halt translation. At the times indicated, cells were lysed and whole-cell lysates immunobloted with anti-GSK3β antibody.
Ijms 08 00051f4
Figure 5. GSK3β exist in a complex with Hsp90. GSK3β was immunoprecipitated from COS cell lysate, the immunoreactive materials were subjected to SDS-PAGE and analyzed by immunoblot using anti-Hsp90 antibody and anti-GSK3β antibidy.
Figure 5. GSK3β exist in a complex with Hsp90. GSK3β was immunoprecipitated from COS cell lysate, the immunoreactive materials were subjected to SDS-PAGE and analyzed by immunoblot using anti-Hsp90 antibody and anti-GSK3β antibidy.
Ijms 08 00051f5

Share and Cite

MDPI and ACS Style

Dou, F.; Chang, X.; Ma, D. Hsp90 Maintains the Stability and Function of the Tau Phosphorylating Kinase GSK3β. Int. J. Mol. Sci. 2007, 8, 51-60. https://doi.org/10.3390/i8010060

AMA Style

Dou F, Chang X, Ma D. Hsp90 Maintains the Stability and Function of the Tau Phosphorylating Kinase GSK3β. International Journal of Molecular Sciences. 2007; 8(1):51-60. https://doi.org/10.3390/i8010060

Chicago/Turabian Style

Dou, Fei, Xingya Chang, and Da Ma. 2007. "Hsp90 Maintains the Stability and Function of the Tau Phosphorylating Kinase GSK3β" International Journal of Molecular Sciences 8, no. 1: 51-60. https://doi.org/10.3390/i8010060

Article Metrics

Back to TopTop