Shu-gang Zhang, Xiao-shan Wang, Ying-dong Zhang, Qing Di Jing-ping Shi Min Qian Li-gang Xu Xing-jian Lin Jie Lu
1 Department of Neurology, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
2 Department of Neurology, Affiliated Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
RESEARCH ARTICLE
Indirubin-3′-monoxime suppresses amyloid-betainduced apoptosis by inhibiting tau hyperphosphorylation
Shu-gang Zhang1,#, Xiao-shan Wang1,#, Ying-dong Zhang1,2,*, Qing Di1, Jing-ping Shi1, Min Qian1, Li-gang Xu1, Xing-jian Lin1, Jie Lu1
1 Department of Neurology, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
2 Department of Neurology, Affiliated Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
Graphical Abstract
#These authors contributed equally to this study.
orcid: 0000-0002-6470-5064 (Ying-dong Zhang)
Accepted: 2016-04-12
Indirubin-3′-monoxime is an effective inhibitor of cyclin-dependent protein kinases, and may play an obligate role in neuronal apoptosis in Alzheimer's disease. Here, we found that indirubin-3′-monoxime improved the morphology and increased the survival rate of SHSY5Y cells exposed to amyloid-beta 25—35 (Aβ25—35), and also suppressed apoptosis by reducing tau phosphorylation at Ser199 and Thr205. Furthermore, indirubin-3′-monoxime inhibited phosphorylation of glycogen synthase kinase-3β (GSK-3β). Our results suggest that indirubin-3′-monoxime reduced Aβ25—35-induced apoptosis by suppressing tau hyperphosphorylation via a GSK-3β-mediated mechanism. Indirubin-3′-monoxime is a promising drug candidate for Alzheimer's disease.
nerve regeneration; indirubin-3′-monoxime; amyloid-beta; Alzheimer's disease; neuronal apoptosis; tau hyperphosphorylation; phosphorylated glycogen synthase kinase-3β; phosphorylated c-Jun N-terminal kinase; neural regeneration
The recognition of pathologic features in patients with Alzheimer's disease (AD) has provided clues to the mechanisms of neuronal apoptosis, and drawn attention to new prospects for AD therapy. A number of experimental models have shown that neuronal death occurs alongside the elicitation of proteins involved in the cell cycle (Lim and Qi, 2003; Colacurcio et al., 2015). Damaged neurons, instead of continuing with the cell cycle after mitosis, initiate abortive processes that result in apoptotic cell death (Xu et al., 2008; Absalon et al., 2013). Cyclin-dependent kinase (CDK) is involved in such processes, and CDK inhibitors might reduce neuronal loss in AD (Zhang et al., 2004; Johnson et al., 2005). There is an urgent need to develop safe, effective, and selective CDK inhibitors that can pass the blood-brain barrier.
Indirubin is a selective CDK inhibitor, which suppresses the activities of CDK1, CDK2, and CDK5. However, it has poor water solubility and liposolubility (Absalon et al., 2013). Indirubin-3′-monoxime (IMX), also a CDK inhibitor, has a low molecular weight and better solubility than indirubin (Zahler et al., 2010; Liao and Leung, 2013). It is nontoxic and acts by competition with adenosine triphosphate at the catalytic site of CDKs (Shelton et al., 2004).The compound was found to suppress tau phosphorylation in Sf9 cells expressing human tau 23 (Leclerc et al., 2001) and, in cerebellar granular neurons, it reduced apoptosis initiated by withdrawal of potassium (Xie et al., 2004). Systemic administration of IMX (20 mg/kg; 3 times per week for 2 months) in APP transgenic mice attenuated spatial memory deficits and decreased presenilin 1 (PS1) mutations in several AD-like phenotypes (Ding et al., 2010).
The aim of the present study was to investigate the neuroprotective effect of IMX against amyloid-beta (Aβ)-induced apoptosis in cultured SH-SY5Y neuroblastoma cells. In addition, we explored the effect of IMX on tau hyperphosphorylation and putative related mechanisms.
SH-SY5Y cell culture
SH-SY5Y cells (Beijing Union Medical College Cell Center, Beijing, China) were grown on RPMI 1640 medium (Gibco BRL, Gaithersburg, MD, USA) supplemented with 15% (v/v) fetal bovine serum (Gibco BRL), 2 mM L-glutamine, 100 μg/mL streptomycin (Sigma, St. Louis, MO, USA), and 100 U/mL penicillin (Sigma) in a humidified atmosphere at 5% CO2and 37°C. The medium was replaced every 2 days; cells were passaged every 3—4 days.
Cell viability assay
Aβ peptide fragment 25—35 (Aβ25—35; Sigma) was dissolved in sterile deionized water and stored at -20°C. IMX (Sigma) was dissolved in dimethyl sulfoxide to 10 mM, stored in aliquots at -20°C, and diluted in medium as necessary. The cells were divided into five groups in a 96-well plate: control (untreated); Aβ (20 μM Aβ25—35); IMX0.2 (0.2 μM IMX + Aβ25—35); IMX0.5 (0.5 μM IMX + Aβ25—35); and IMX1 (1.0 μM IMX + Aβ25—35). Cells in the Aβ groups were incubated with Aβ25—35at 37°C for 7 days. Cells in the IMX groups were incubated at 37°C for 2 days. We used 2-(2-methoxy-4-nitrophenyl)-3-(4-nitrophenyl)-5-(2,4-disulfophenyl)-2H tetrazolium (WST-8, Cell Counting Kit-8; Dojindo, Kumamoto, Japan; Tsukatani et al., 2011) to examine the effect of IMX on Aβ25—35-induced changes in SH-SY5Y cells. After treatment, the medium was replaced by Dulbecco's modified Eagle's medium to stop the effects of IMX and Aβ25—35. WST-8 (10 μL) was added to each well, and the plate was incubated for 4 hours. Absorbance was measured at 450 nm using a Dynatech MR5000 reader (BMG Labtech, Offenburg, Germany) with a reference wavelength of 630 nm.
Flow cytometry
To determine the effect of IMX on early and late apoptosis and necrosis triggered by Aβ25—35, we examined SH-SY5Y cells using flow cytometry. The cells were transferred into six-well plates and incubated with IMX (0, 0.5, and 1.0 μM) for 24 hours. Aβ25—35(20 μM) was then added and mixed. Forty-eight hours later, the cells that remained fixed to the plates were collected in phosphate buffered saline (PBS) and combined with the floating dead cells. Cells (approximately 1 × 106) were washed twice with cold PBS and resuspended in 200 μL cold 1× binding buffer. Annexin V—FITC (10 μL) and propidium iodide (PI; 5 μL) were admixed and incubated in the dark at room temperature for 15 minutes, then 300 μL binding buffer was added. Cells were quantified instantly in a flow cytometer (FACSCalibur, Becton Dickinson Immunocytometry Systems, San Jose, CA, USA), using emission filters at 525 and 575 nm. Cells negative for both annexin V and PI (V-/PI-) were considered normal, whereas a V+/PI-result was considered a criterion of early apoptosis, and V+/ PI+was assumed to indicate late apoptotic-like cell death and necrosis. A minimum of 10,000 cells per experiment were assayed in the three separate trials.
Western blot assay
To evaluate the effects of Aβ25—35and IMX on the phosphorylation of tau, glycogen synthase kinase 3β (GSK-3β) and c-Jun N-terminal kinase (JNK) in SH-SY5Y cells, the cells were incubated with a mixture of 20 μM Aβ25—35and 0.5 μM IMX for 6 hours. They were then washed twice with cold PBS, and lysed in cell lysis buffer for 30 minutes on ice. The soluble portion was produced by centrifugation (15,000 × g for 15 minutes at 4°C). Protein concentrations were established using the bicinchoninic acid method (Yalamati et al., 2015) (Pierce Biotechnology, Rockford, IL, USA). Aliquots of total protein were boiled for 10 minutes in loading buffer and subsequently separated in 10% sodium dodecyl sulfate-polyacrylamide gel. Next, the proteins were transferred onto nitrocellulose membranes by electroporation (Immobilon TMP, Millipore Corp., Bedford, MA, USA) using a Trans-Blot system (Bio-Rad, New York, NY, USA). The membranes were then blocked with 5% nonfat milk in Tris-buffered saline with Tween-20 (TTBS; 10 mM Tris-HCl, 150 mM NaCl, 0.2% Tween-20) for 1 hour at room temperature. Samples were incubated with monoclonal primary antibodies (β-actin, 1:10,000, Sigma; polyclonal anti-tau pS199, 1:1,000, BioSource Int., Camarillo, CA, USA; anti-tau pT205, 1:1,000, Bioworld Technology (St. Louis Park, MN, USA); Ser-9-phosphorylated GSK-3β (p-GSK-3β Ser9), phosphorylated JNK (p-JNK), and JNK, all 1:1,000; Cell Signaling Technology, Beverly, MA, USA) at 4°C overnight. The membranes were washed twice with TTBS, and incubated with the secondary antibody (anti-rabbit-horseradish peroxidase, 1:5,000; Cell Signaling Technology) at room temperature for 1 hour. Proteins were then visualized using the ECL Advanced Western Blotting Detection kit (Amersham Biosciences Ltd., Amersham, UK), and the mean optical density of each band was calculated using a Fluor-S MultiImager (Bio-Rad Laboratories (Shanghai) Co., Ltd., Shanghai, China) with Quantity One software (Bio-Rad Laboratories (Shanghai) Co., Ltd.).
Statistical analysis
The data, presented as the mean ± SEM, were analyzed using SPSS 11.0 software (SPSS Inc., Chicago, IL, USA). Means were compared by one-way analysis of variance and the least significant difference post hoc test. P < 0.05 was considered statistically significant.
Figure 1 Effect of IMX on viability of SH-SY5Y cells exposed to Aβ25-35.
Figure 2 Apoptosis triggered by Aβ25-35was suppressed by IMX (flow cytometry).
IMX enhanced the viability of SH-SY5Y cells exposed to Aβ25-35
Under an inverted phase contrast microscope (Olympus Optical Co., Ltd., Tokyo, Japan), SH-SY5Y cells in the control group appeared well-formed than those in the Aβ group. Pretreatment with different concentrations of IMX markedly improved morphology after Aβ25—35exposure (Figure 1A).
Cell viability after Aβ25—35exposure was significantly lower than in control cells (P < 0.001). However, pretreatment with IMX (0.5 μM and 1.0 μM) increased cell viability in a concentration-dependent manner (P < 0.01; Figure 1B).
IMX reduced neuronal apoptosis triggered by Aβ25-35in SH-SY5Y cells
Flow cytometry showed that there were more apoptotic cells after exposure to 20 μM Aβ25—35than in the control group (P < 0.01), but significantly fewer in the IMX group (12.4 ± 1.82%) than in the Aβ group (20.33 ± 2.02%; P < 0.01). This indicates that IMX protected cells against Aβ-induced apoptosis (Figure 2).
IMX decreased tau phosphorylation caused by Aβ25-35
Western blot assay revealed significantly more tau phosphorylation at Ser199 and Thr205 in the Aβ group than in the control group (P < 0.05). This effect was markedly reduced by co-treatment with IMX at 0.5 μM and 1.0 μM (P < 0.05; Figure 3).
Effects of IMX on p-GSK-3β expression
p-GSK-3β (Ser9) expression was lower after Aβ25—35exposure than in control cells, indicating that GSK-3β activity was increased. However, p-GSK-3β (Ser9) was markedly overexpressed after pretreatment with IMX (P < 0.05; Figure 4A). These results suggest that GSK-3β is involved in the effect of IMX on Aβ-induced tau phosphorylation.
IMX treatment did not affect p-JNK expression
The expression of p-JNK in the Aβ group was markedly higher than that in the control group (P < 0.05). However, there were no significant changes after IMX treatment (Figure 4B), indicating that p-JNK is not involved in the effect of IMX on Aβ-induced tau phosphorylation.
Figure 3 IMX decreased Aβ25-35-induced tau phosphorylation at Ser (A) and Thr205 (B).
Figure 4 Effect of IMX on p-GSK-3β (A) and p-JNK (B) expression in the presence of Aβ25-35.
SH-SY5Y neuroblastoma cells are a well-characterized human cell model for investigating the pharmacological effects of IMX. In the present study, we exposed the cells to 20 μM Aβ25—35, which showed a low level of neurotoxicity. Aggregated Aβ25—35reduced cell viability and initiated apoptosis. Notably, our results provide evidence for the dose-dependent protective effect of IMX against Aβ-induced cell death, consistent with the findings of our previous investigation (Zhang et al., 2009).
Aβ induces and maintains the pathogenic changes in AD, but tau protein also has an important role in the progression of the disease (Iqbal et al., 2014). Aβ amyloidosis triggers the starting phase of tau accumulation and phosphorylation at Ser199, Thr231, and Ser396 in APP Sw mice (Tomidokoro et al., 2001; Stein et al., 2004). In cultured Neuro-2a cells (Hu et al., 2004; Jung et al., 2012; Huang et al., 2014; Nicole et al., 2014; Deng et al., 2015; Zhang et al., 2015), SH-SY5Y cells, and hippocampal neurons (Lafay-Chebassier et al., 2005; Jin et al., 2011; Reifert et al., 2011; Doherty et al., 2013; Martins et al., 2013; Sui et al., 2015), Aβ markedly diminished cell viability, increased the number of apoptotic-like cells, and promoted tau phosphorylation. Inhibiting tau phosphorylation has become a viable approach to treating or even preventing AD. Here, Aβ25—35exposure elevated the rate of tau phosphorylation at pS199 and pT205 in serum-free cultured SH-SY5Y cells, supporting the findings of a previous study (Sun et al., 2008). We also found that IMX suppressed tau phosphorylation induced by Aβ25—35. This suggests that the effect of IMX on Aβ25—35-induced neurotoxicity may be via the inhibition of tau phosphorylation.
GSK-3β, also called Tau Protein Kinase I, is a proline-directed serine/threonine kinase, which phosphorylates tau at a number of AD-relevant epitopes in vitro and in transfected cells (Hanger et al., 1992; Ishiguro et al., 1992, 1993; Mandelkow et al., 1992; Mulot et al., 1994; Sperbera et al., 1995;Bass et al., 2015). GSK-3β may have a pivotal function in the relationship between Aβ peptides and phosphorylated tau, and triggers a pathogenic cycle in AD (Wang et al., 2006; Dobarro et al., 2013; Hoppe et al., 2013; Xian et al., 2014; Amin et al., 2015; Kim et al., 2015; Vossel et al., 2015), resulting in a more important effect than those caused by other kinases such as cdk/MAPK (Elyaman et al., 2002; Liu et al., 2002). The activation of GSK-3β is reportedly related to its low rate of phosphorylation at residue Ser9 (Stambolic and Woodgett, 1994; Wang et al., 1994; Murai et al., 1996). In the present study, we used an antibody against p-GSK-3β Ser9 to investigate the activation of GSK-3β in SH-SY5Y cells. After exposure to Aβ25—35, p-GSK-3β (Ser9) expression was decreased, indicating that GSK-3β activity was elevated. However, pretreatment with IMX led to a considerable rise in the expression of p-GSK-3β (Ser9), which suggests that GSK-3β contributes critically to the action of IMX on Aβ-initiated tau phosphorylation.
The constituents of the mitogen-activated protein kinase (MAPK) family, including extracellular signal-regulated kinase, p38 MAPK, and JNK, are enzymes of major importance in the hyperphosphorylation of tau. Because the activation of JNK is critically involved in Aβ-induced cell death (Wei et al., 2002), we investigated the effects of IMX on JNK activation after Aβ25—35exposure. Expression of p-JNK in the Aβ25—35-exposed cells was significantly higher than in control cells. However, there were no significant changes after IMX treatment, indicating that IMX did not influence JNK in our cell system.
In summary, IMX exerted neuroprotective effects by preventing Aβ-induced damage, via a mechanism that likely involves inhibition of tau phosphorylation. The suppression of GSK-3β signaling was the most important route by which IMX suppressed phosphorylation. Taking into consideration the important role of Aβ throughout the pathogenesis of AD, our results suggest that IMX is a promising drug candidate for the treatment of AD.
Author contributions: YDZ, SGZ and XSW conceived and designed the study. XJL, SGZ, LGX and JL performed the experiments. SGZ, JPS, MQ and QD analyzed the data and wrote the paper. All authors approved the final version of the paper.
Conflicts of interest: None declared.
Plagiarism check: This paper was screened twice using Cross-Check to verify originality before publication.
Peer review: This paper was double-blinded and stringently reviewed by international expert reviewers.
Absalon S, Kochanek DM, Raghavan V, Krichevsky AM (2013) MiR-26b, upregulated in Alzheimer's disease, activates cell cycle entry, tau-phosphorylation, and apoptosis in postmitotic neurons. J Neurosci 33:14645-14659.
Amin J, Paquet C, Baker A, Asuni AA, Love S, Holmes C, Hugon J, Nicoll JA, Boche D (2015) Effect of amyloid-β (Aβ) immunization on hyperphosphorylated tau: a potential role for glycogen synthase kinase (GSK)-3β. Neuropathol Appl Neurobiol 41:445-457.
Bass B, Upson S, Roy K, Montgomery EL, Jalonen TO, Murray IV (2015) Glycogen and amyloid-beta: key players in the shift from neuronal hyperactivity to hypoactivity observed in Alzheimer's disease?. Neural Regen Res 10:1023-1025.
Colacurcio DJ, Zyskind JW, Jordan-Sciutto KL, Espinoza CA (2015) Caspase-dependent degradation of MDMx/MDM4 cell cycle regulatory protein in amyloid β-induced neuronal damage. Neurosci Lett 609:182-188.
Deng J, Habib A, Obregon DF, Barger SW, Giunta B, Wang YJ, Hou H, Sawmiller D, Tan J (2015) Soluble amyloid precursor protein alpha inhibits tau phosphorylation through modulation of GSK3β signaling pathway. J Neurochem 135:630-637.
Ding Y, Qiao A, Fan GH (2010) Indirubin-3'-monoxime rescues spatial memory deficits and attenuates β-amyloid-associated neuropathology in a mouse model of Alzheimer's disease. Neurobiol Dis 39:156-168.
Dobarro M, Gerenu G, Ramírez MJ (2013) Propranolol reduces cognitive deficits, amyloid and tau pathology in Alzheimer's transgenic mice. Int J Neuropsychopharmacol 16:2245-2257.
Doherty GH, Beccano-Kelly D, Yan SD, Gunn-Moore FJ, Harvey J (2013) Leptin prevents hippocampal synaptic disruption and neuronal cell death induced by amyloid β. Neurobiol Aging 34:226-237.
Elyaman W, Yardin C, Hugon J (2002) Involvement of glycogen synthase kinase-3beta and tau phosphorylation in neuronal Golgi disassembly. J Neurochem 81:870-880.
Hanger DP, Hughes K, Woodgett JR, Brion JP, Anderton BH (1992) Glycogen synthase kinase-3 induces Alzheimer's disease-like phosphorylation of tau: Generation of paired helical filament epitopes and neuronal localisation of the kinase. Neurosci Lett 147:58-62.
Hoppe JB, Coradini K, Frozza RL, Oliveira CM, Meneghetti AB, Bernardi A, Pires ES, Beck RCR, Salbego CG (2013) Free and nanoencapsulated curcumin suppress β-amyloid-induced cognitive impairments in rats: involvement of BDNF and Akt/GSK-3β signaling pathway. Neurobiol Learn Mem 106:134-144.
Hu J, Geng M, Li J, Xin X, Wang J, Tang M, Zhang J, Zhang X, Ding J (2004) Acidic oligosaccharide sugar chain, a marine-derived acidic oligosaccharide, inhibits the cytotoxicity and aggregation of amyloid beta protein. J Pharmacol Sci 95:248-255.
Huang HC, Tang D, Xu K, Jiang ZF (2014) Curcumin attenuates amyloid-β-induced tau hyperphosphorylation in human neuroblastoma SH-SY5Y cells involving PTEN/Akt/GSK-3β signaling pathway. J Recept Signal Transduct Res 34:26-37.
Iqbal K, Kazim SF, Bolognin S, Blanchard J (2014) Shifting balance from neurodegeneration to regeneration of the brain: a novel therapeutic approach to Alzheimer's disease and related neurodegenerative conditions. Neural Regen Res 9:1518-1519.
Ishiguro K, Omori A, Takamatsu M, Sato K, Arioka M, Uchida T, Imahori K (1992) Phosphorylation sites on tau by tau protein kinase I, a bovine derived kinase generating an epitope of paired helical filaments. Neurosci Lett 148:202-206.
Ishiguro K, Shiratsuchi A, Sato S, Omori A, Arioka M, Kobayashi S, Uchida T, Imahori K (1993) Glycogen synthase kinase 3 beta is identical to tau protein kinase I generating several epitopes of paired helical filaments. FEBS Lett 325:167-172.
Jin M, Shepardson N, Yang T, Chen G, Walsh D, Selkoe DJ (2011) Soluble amyloid β-protein dimers isolated from Alzheimer cortex directly induce Tau hyperphosphorylation and neuritic degeneration. Proc Natl Acad Sci U S A 108:5819-5824.
Johnson K, Liu L, Majdzadeh N, Chavez C, Chin PC, Morrison B, Wang L, Park J, Chugh P, Chen HM, D'Mello SR (2005) Inhibition of neuronal apoptosis by the cyclin-dependent kinase inhibitor GW8510: identification of 3' substituted indolones as a scaffold for the development of neuroprotective drugs. J Neurochem 93:538-548.
Jung KM, Astarita G, Yasar S, Vasilevko V, Cribbs DH, Head E, Cotman CW, Piomelli D (2012) An amyloid β(42)-dependent deficit in anandamide mobilization is associated with cognitive dysfunction in Alzheimer's disease. Neurobiol Aging 33:1522-1532.
Kim HG, Park G, Lim S, Park H, Choi JG, Jeong HU, Kang MS, Lee MK, Oh MS (2015) Mori Fructus improves cognitive and neuronal dysfunction induced by beta-amyloid toxicity through the GSK-3β pathway in vitro and in vivo. J Ethnopharmacol 171:196-204.
Lafay-Chebassier C, Paccalin M, Page G, Barc-Pain S, Perault-Pochat MC, Gil R, Pradier L, Hugon J (2005) mTOR/p70S6k signalling alteration by Abeta exposure as well as in APP-PS1 transgenic models and in patients with Alzheimer's disease. J Neurochem 94:215-225.
Leclerc S, Garnier M, Hoessel R, Marko D, Bibb JA, Snyder GL, Greengard P, Biernat J, Wu YZ, Mandelkow EM, Eisenbrand G, Meijer L (2001) Indirubins inhibit glycogen synthase kinase-3 beta and CDK5/p25, two protein kinases involved in abnormal tau phosphorylation in Alzheimer's disease. A property common to most cyclin-dependent kinase inhibitors? J Biol Chem 276:251-260.
Liao XM, Leung KN (2013) Indirubin-3'-oxime induces mitochondrial dysfunction and triggers growth inhibition and cell cycle arrest in human neuroblastoma cells. Oncol Rep 29:371-379.
Lim AC, Qi RZ (2003) Cyclin-dependent kinases in neural development and degeneration. J Alzheimers Dis 5:329-335.
Liu F, Iqbal K, Grundke-Iqbal I, Gong CX (2002) Involvement of aberrant glycosylation in phosphorylation of tau by cdk5 and GSK-3beta. FEBS Lett 530:209-214.
Mandelkow EM, Drewes G, Biernat J, Gustke N, Van Lint J, Vandenheede JR, Mandelkow E (1992) Glycogen synthase kinase-3 and the Alzheimer-like state of microtubule-associated protein tau. FEBS Lett 314:315-321.
Martins I, Gomes S, Costa RO, Otvos L, Oliveira CR, Resende R, Pereira CMF (2013) Leptin and ghrelin prevent hippocampal dysfunction induced by Aβ oligomers. Neuroscience 241:41-51.
Mulot SF, Hughes K, Woodgett JR, Anderton BH, Hanger DP (1994) PHF-tau from Alzheimer's brain comprises four species on SDSPAGE which can be mimicked by in vitro phosphorylation of human brain tau by glycogen synthase kinase-3 beta. FEBS Lett 349:359-364.
Murai H, Okazaki M, Kikuchi A (1996) Tyrosine dephosphorylation of glycogen synthase kinase-3 is involved in its extracellular signal-dependent inactivation. FEBS Lett 392:153-160.
Nicole R, Giulia Di B, Carmela P, Salvatore A, Renato B, Giuseppina C (2014) CHF5074 protects SH-SY5Y human neuronal-like cells from amyloidbeta 25-35 and tumor necrosis factor related apoptosis inducing ligand toxicity in vitro. Curr Alzheimer Res 11:714-724.
Reifert J, Hartung-Cranston D, Feinstein SC (2011) Amyloid beta-mediated cell death of cultured hippocampal neurons reveals extensive Tau fragmentation without increased full-length tau phosphorylation. J Biol Chem 286:20797-20811.
Shelton SB, Krishnamurthy P, Johnson GV (2004) Effects of cyclin-dependent kinase-5 activity on apoptosis and tau phosphorylation in immortalized mouse brain cortical cells. J Neurosci Res 76:110-120.
Sperbera BR, Leight S, Goedert M, Lee VMY (1995) Glycogen synthase kinase-3β phosphorylates tau protein at multiple sites in intact cells. Neurosci Lett 197:149-153.
Stambolic V, Woodgett JR (1994) Mitogen inactivation of glycogen synthase kinase-3 beta in intact cells via serine 9 phosphorylation. Biochem J 303:701-704.
Stein TD, Anders NJ, DeCarli C, Chan SL, Mattson MP, Johnson JA (2004) Neutralization of transthyretin reverses the neuroprotective effects of secreted amyloid precursor protein (APP) in APPSW mice resulting in tau phosphorylation and loss of hippocampal neurons: support for the amyloid hypothesis. J Neurosci 24:7707-7717.
Sui HJ, Zhang LL, Liu Z, Jin Y (2015) Atorvastatin prevents A[beta] oligomer-induced neurotoxicity in cultured rat hippocampal neurons by inhibiting Tau cleavage. Acta Pharmacol Sin 36:553-564.
Sun ZK, Yang HQ, Pan J, Zhen H, Wang ZQ, Chen SD, Ding JQ (2008) Protective effects of erythropoietin on tau phosphorylation induced by beta-amyloid. J Neurosci Res 86:3018-3027.
Tomidokoro Y, Ishiguro K, Harigaya Y, Matsubara E, Ikeda M, Park JM, Yasutake K, Kawarabayashi T, Okamoto K, Shoji M (2001) Aβ amyloidosis induces the initial stage of tau accumulation in APPSw mice. Neurosci Lett 299:169-172.
Tsukatani T, Suenaga H, Higuchi T, Shiga M, Noguchi K, Matsumoto K (2011) Distinction of Gram-positive and -negative bacteria using a colorimetric microbial viability assay based on the reduction of water-soluble tetrazolium salts with a selection medium. J Gen Appl Microbiol 57:331-339.
Vossel KA, Xu JC, Fomenko V, Miyamoto T, Suberbielle E, Knox JA, Ho K, Kim DH, Yu GQ, Mucke L (2015) Tau reduction prevents Aβ-induced axonal transport deficits by blocking activation of GSK3β. J Cell Biol 209:419-433.
Wang QM, Fiol CJ, DePaoli-Roach AA, Roach PJ (1994) Glycogen synthase kinase-3 beta is a dual specificity kinase differentially regulated by tyrosine and serine/threonine phosphorylation. J Biol Chem 269:14566-14574.
Wang ZF, Li HL, Li XC, Zhang Q, Tian Q, Wang Q, Xu H, Wang JZ (2006) Effects of endogenous β-amyloid overproduction on tau phosphorylation in cell culture. J Neurochem 98:1167-1175.
Wei W, Wang X, Kusiak JW (2002) Signaling events in amyloid beta-peptide-induced neuronal death and insulin-like growth factor I protection. J Biol Chem 277:17649-17656.
Xian YF, Mao QQ, Wu JC, Su ZR, Chen JN, Lai XP, Ip SP, Lin ZX (2014) Isorhynchophylline treatment improves the amyloid-β-induced cognitive impairment in rats via inhibition of neuronal apoptosis and tau protein hyperphosphorylation. J Alzheimers Dis 39:331-346.
Xie Y, Liu Y, Ma C, Yuan Z, Wang W, Zhu Z, Gao G, Liu X, Yuan H, Chen R, Huang S, Wang X, Zhu X, Wang X, Mao Z, Li M (2004) Indirubin-3′-oxime inhibits c-Jun NH2-terminal kinase: anti-apoptotic effect in cerebellar granule neurons. Neurosci Lett 367:355-359.
Xu L, Di Q, Zhang Y (2008) Cell cycle proteins preceded neuronal death after chronic cerebral hypoperfusion in rats. Neurol Res 30:932-939.
Yalamati P, Bhongir AV, Karra M, Beedu SR (2015) Comparative Analysis of Urinary Total Proteins by Bicinchoninic Acid and Pyrogallol Red Molybdate Methods. J Clin Diagn Res 9:BC01-BC04.
Zahler S, Liebl J, Fürst R, Vollmar AM (2010) Anti-angiogenic potential of small molecular inhibitors of cyclin dependent kinases in vitro. Angiogenesis 13:239-249.
Zhang LF, Zhou ZW, Wang ZH, Du YH, He ZX, Cao C, Zhou SF (2015) Coffee and caffeine potentiate the antiamyloidogenic activity of melatonin via inhibition of Aβ oligomerization and modulation of the Tau-mediated pathway in N2a/APP cells. Drug Des Devel Ther 9:241-272.
Zhang M, Li J, Chakrabarty P, Bu B, Vincent I (2004) Cyclin-dependent kinase inhibitors attenuate protein hyperphosphorylation, cytoskeletal lesion formation, and motor defects in Niemann-Pick Type C mice. Am J Pathol 165:843-853.
Zhang S, Zhang Y, Xu L, Lin X, Lu J, Di Q, Shi J, Xu J (2009) Indirubin-3′-monoxime inhibits β-amyloid-induced neurotoxicity in neuroblastoma SH-SY5Y cells. Neurosci Lett 450:142-146.
Copyedited by Slone-Murphy J, Hindle A, Yu J, Qiu Y, Li CH, Song LP, Zhao M
10.4103/1673-5374.184500
How to cite this article: Zhang SG, Wang XS, Zhang YD, Di Q, Shi JP, Qian M, Xu LG, Lin XJ, Lu J (2016) Indirubin-3′-monoxime suppresses amyloid-beta-induced apoptosis by inhibiting tau hyperphosphorylation. Neural Regen Res 11(6)∶988-993.
Funding: This research was supported by the Nanjing Medical Science and Technique Development Foundation of China, No. QRX11199; a grant from the Nanjing Science and Technology Commission Project of China, No. 201303010; and a grant from the Health Research Project in Nanjing City of China, No. YKK14101.
*Correspondence to: Ying-dong Zhang, M.D., zhangyingdong@aliyun.com.