Formononetin Protects Neurons Against Hypoxia-Induced Cytotoxicity ...

4 downloads 0 Views 1MB Size Report
dDepartment of Neurology, Peking University Third Hospital, Beijing, China. 11. Handling Associate Editor: Chengxin Gong. 12. Accepted 16 October 2011.
1

Journal of Alzheimer’s Disease 28 (2011) 1–14 DOI 10.3233/JAD-2011-110506 IOS Press

3

roo f

2

Formononetin Protects Neurons Against Hypoxia-Induced Cytotoxicity Through Upregulation of ADAM10 and sA␤PP␣

or P

1

6

Miao Suna,1 , Ting Zhoua , Liang Zhoua,1 , Qiang Chenb,∗ , Yan Yua , Huan Yanga , Kaiyin Zhonga , Ximeng Zhanga , Feng Xuc , Shaoqing Caic , Albert Yua , Hui Zhangd , Ruizhong Xiaod , Dongsheng Xiaod and Dehua Chuia,d,∗

7

a Neuroscience

4 5

12

Handling Associate Editor: Chengxin Gong

dA

9 10

uth

11

Research Institute and Department of Neurobiology, Key Laboratory for Neuroscience, Ministry of Education and Ministry of Public Health, Health Science Center, Peking University, Beijing, China b Department of Neurology, Ningbo Beilun Hospitals, Ningbo, China c Department of Natural Medicines, School of Pharmaceutical Sciences, Peking University, Beijing, China d Department of Neurology, Peking University Third Hospital, Beijing, China

8

cte

Accepted 16 October 2011

24

Keywords: ADAM10, amyloid-␤ protein precursor (A␤PP), formononetin, soluble-A␤PP␣

25

Supplementary data available online: http://www.j-alz.com/issues/28/vol28-4.html#supplementarydata03

16 17 18 19 20 21 22

co

15

Un

14

rre

23

Abstract. Formononetin, an active constituent of the Chinese herb Astragali Radix, has been reported to have beneficial effects for Alzheimer’s disease (AD). Yet the mechanism of this effect remains to be elucidated. The present study shows that formononetin increases soluble-A␤PP␣ (sA␤PP␣) secretion and thus protects human-A␤PP Swedish mutation cell (N2a-A␤PP cell) from hypoxia-induced apoptosis. Using hypoxic N2a-A␤PP cell as an in vitro model of AD-like pathology, we confirmed that regular treatment with formononetin could have neuroprotective effects, followed respectively by reduced caspase 3 activity and increased cell viability. Strikingly, our data revealed that the caspase 3-blocking effect of formononetin was largely mediated by stimulation of ␣-secretase cleavage of A␤PP, and increasing the secretion of its soluble form, sA␤PP␣. Moreover, the protective effect of formononetin was totally inhibited by TAPI-2, an ␣-secretase complex inhibitor, suggesting the role of the sA␤PP␣ pathway in the neuroprotective response to formononetin. We also found that the stimulative effect of formononetin on ␣-secretase activity was mainly conducted by upregulating ADAM10 expression at the transcriptional level. Altogether, our study provides novel insights into how formononetin mediates stimulation of the ADAM10-sA␤PP␣ pathway and exerts a neuronal protective effect.

13

INTRODUCTION

1 These

authors contributed equally to this paper. ∗ Correspondence to: Dr. Dehua Chui, Neuroscience Research Institute, Peking University Health Science Center, 38 Xueyuan Road, Hai Dian District, 100191, Beijing, China. E-mail: [email protected] or Dr. Qiang Chen, Department of Neurology, Ningbo Beilun Hospitals, Ningbo 315806, China.

Formononetin [7-hydroxy-3 (4-methoxypheny) chromone or 4 -methoxy daidzein] is a soy isoflavonoid found abundantly in traditional Chinese medicine Astragalus mongholicus (Bunge) and Trifolium pretense L. (red clover). It belongs to the phytoestrogens due to its similar chemical structure to gonadal steroid estrogen [1]. Studies have found

ISSN 1387-2877/11/$27.50 © 2011 – IOS Press and the authors. All rights reserved

26

27 28 29 30 31 32 33

MATERIALS AND METHODS

72

Formononetin extraction

40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65 66 67 68 69

73 74 75 76 77 78 79 80 81

cte

39

rre

38

co

37

Un

36

Fig. 1. Structure of formononetin. 13 C-MNR.

or P

71

35

The purity of formononetin by HPLC-UV analysis was above 98% (Fig. 1). Reagents

3-[4,5-Dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide (MTT), sodium dodecyl sulfate (SDS), and N, N-dimethyl sulfoxide (DMSO), were purchased from Sigma (St. Louis, MO, USA). The ␣-secretase inhibitor TAPI-2 was purchased from Calbiochem (San Diego, CA, USA). Dulbecco’s modified Eagle’s medium (DMEM) and fetal bovine serum (FBS) were obtained from Invitrogen (Carlsbad, CA, USA). The LDH kit was purchased from Promega (Madison, WI, USA), and the sA␤PP␣ activity kit was purchased from R&D Systems (Minneapolis, MN, USA).

dA

70

that formononetin can promote endothelial repair and wound healing [2], prevent lipopolysaccharideinduced injury in dopaminergic neurons [3], and protect PC12 cells as well as cultured cortical and hippocampal neurons from oxidative stress and toxicity induced by L-glutamate or amyloid-␤ [4–7], suggesting a neuroprotective effect of formononetin. The soluble fragment of the amyloid-␤ protein precursor (sA␤PP␣) is the product of A␤PP cleavage by ␣-secretase in the non-amyloidogenic pathway. It has been suggested that decreased sA␤PP␣ correlates with memory loss during aging in a rat model [8], and generation of sA␤PP␣ has been found to be down-regulated in aging human fibroblasts [9]. Moreover, sA␤PP␣ formation tends to be down-regulated in response to ischemia [10, 11]. In neuronal precursors and nonneuronal cells, sA␤PP␣ acts as a proliferative factor [12–14], involved in both cell differentiation induction [15] and neurite outgrowth [16]. Besides these proliferative effects, sA␤PP␣ also plays an important role in neuronal plasticity and memory [17, 18]. Furthermore, sA␤PP␣ has been found to have a neuroprotective effect against cell apoptosis [20] and to improve the outcome of severe brain injury [19]. Therefore upregulating sA␤PP␣ could be a potential target for AD treatment, and to find out whether and how formononetin could regulate A␤PP processing and sA␤PP␣ formation would be of great interest. Here, we used a mouse neuroblastoma cell line stably transfected with human-A␤PP Swedish mutation (N2a-A␤PP) to evaluate the effects of formononetin on hypoxia-induced reduction of sA␤PP␣ secretion and subsequent neuron injury. Results of the study provide a novel neuroprotective mechanism of formononetin mediated by increasing sA␤PP secretion and ␣secretase and A-Disintegrin-And-Metalloproteinase (ADAM) 10 expression.

34

roo f

M. Sun et al. / Formononetin Increases sA␤PP␣ and ADAM10 Level

uth

2

Astragali Radix was collected from Hunyuan County, Shanxi Province of China, and authenticated by Professor Shao-Qing CAI as the roots of Astragalus membranaceus (Fisch.) Bge. var. mongholicus 11/4/2011(Bge.) Hsiao. Formononetin was isolated from Astragali Radix by chromatographic methods, including column chromatography on D-101 macroporous adsorption resin, silica gel, and was identified on the basis of spectral data, including UV, 1 H-NMR, and

Cell culture and hypoxia treatments N2a cells stably transfected with humanA␤PP695sw (N2a-A␤PP) were grown in DMEM supplemented with 10% FBS, at 37◦ C in an atmosphere of 5% CO2 . For formononetin and hypoxia treatment, cells were pre-treated with formononetin for 4 h and then transferred into an anaerobic chamber (model 1029, Forma Scientific, Marietta, OH, USA, [21]) containing 85% N2 /10% H2 /5% CO2 , for various durations. Cell viability assay MTT assay was carried out as previously described [22]. Cells were plated in 96-well plates, and the cell viability was determined by the conventional MTT reduction assay. The cells were exposed to hypoxic condition at different time points with/without various concentrations of formononetin for the indicated time. After incubation, cells were treated with the MTT solution (0.5 mg/ml) for 4 h at 37◦ C in the dark. 200 ␮l DMSO was added to the culture medium and mixed with a pipette until the blue formazan dissolved completely. The optical density of formazan was measured at 570 nm using a micro plated reader

82 83

84

85 86 87 88 89 90 91 92 93 94 95

96

97 98 99 100 101 102 103 104 105

106

107 108 109 110 111 112 113 114 115 116 117 118

M. Sun et al. / Formononetin Increases sA␤PP␣ and ADAM10 Level

124 125 126 127 128 129 130 131 132 133 134 135 136 137 138

139

As the key index for apoptosis detection, Annexin V-PI (Sigma) staining was performed to measure phosphatidylserine externalization in N2a-A␤PP cells. Briefly, cells were processed with different treatments, then washed twice with PBS and resuspended in 200 ␮l of binding buffer (10 mM HEPES, pH 7.4, 140 mM NaCl, 1 mM MgCl2 , 5 mM KCl, 2.5 mMCaCl2 ). 10 ␮l FITC-conjugated AnnexinV was then added to give a final concentration of 0.5 ␮g/ml. The staining sample was incubated at room temperature for 20 min, without light. Subsequently, 5 ␮l of propidium iodine was added to the samples (final concentration of 1 ␮g/ml) and 10,000 cells were immediately analyzed using a FACSCalibur flow cytometer (Becton Dickinson). Results were calculated as a percentage of apoptotic cells. Lactate dehydrogenase release assay

162

α-secretase activity measurement

144 145 146 147 148 149 150 151 152 153 154 155 156 157 158 159 160

163 164

rre

143

co

142

Un

141

Immunoprecipitation, electrophoresis, and Western blot analysis Western blotting and immunoprecipitaion assay were performed as previously described with minor modifications [23]. After the indicated treatments, conditioned medium was collected, mixed with the complete protease inhibitor cocktail (Roche), and centrifuged at 3,000 g for 10 min to remove cell debris. Cells were harvested and lysed on ice in Western blot lysis buffer containing 50 mM Tris-HCl, pH 6.8, 8 M urea, 5% ␤-mercaptoethanol, 2% SDS, and protease inhibitors. The lysates were collected, centrifuged at 12,000 g for 5 min, and quantified for total proteins by the BCA protein assay kit. To detect sA␤PP␣, equivalent volumes of conditioned medium were directly analyzed by 10% sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE), based on the protein concentration of cells in each plate. sA␤PP␣ were immunoprecipitated from conditioned medium using a monoclonal A␤specific antibody 6E10 as described previously [24], and culture supernatants were normalized for total cellular protein to correct for variations in cell number. For analysis of flA␤PP, and CTF␣, cell lysates were separated on 10% T 5% C Bicine/Tris, 8 M urea, SDS-PAGE [25]. For western blot analysis, protein was transferred to 0.45 ␮m polyvinylidene difluoride membranes (Immobilon-P; Millipore, Bedford, MA, USA), blocked for 1 h with 5% (m/v) nonfat milk in Tris-buffered saline (pH 7.5), supplemented with 0.1% Tween 20. Antibodies and their dilutions used in this study include A8717 (1 : 10000) for A␤PP derivatives and anti-␤-actin mouse monoclonal antibody (1 : 5,000) as an internal reference control. For ADAMs analysis, cells were harvested on ice, homogenized, and suspended in RIPA buffer (50 mM Tris-HCl, pH 8, 150 mM NaCl, 0.1% SDS, 1% Nonidet P-40, 0.5% sodium deoxycholate, and complete protease inhibitor cocktail). Extracts (60 ␮g of protein) were subjected to electrophoresis, and separated

cte

161

Lactate dehydrogenase (LDH) is a soluble cytosolic enzyme present in most eukaryotic cells, released into the culture medium upon cell death due to the damage of plasma membrane. The increase in LDH activity in the culture supernatant is proportional to the number of lysed cells. After cells were exposed to hypoxia in the absence or presence of formononetin for 18 h, the supernatant and cell lysates were collected, and the amount of LDH release was determined using an assay kit according to the manufacturer’s protocol. Briefly, the supernatant and cell lysates were transferred to 96-well plates and incubated with 1 mg/ml NADH in pyruvate substrate solution at 37◦ C for 15 min. After additional incubation at 37◦ C for 15 min with 2, 4dinitrophenylhydrazine, the reaction was stopped by addition of 0.4 M NaOH. The changes in absorbance were determined at 440 nm using a spectrophotometer microplate reader. LDH leakage was expressed as the percentage (%) of the total LDH activity (LDH in the supernatant + LDH in the cell lysate), according to the equation: %LDH released = (LDH activity in the medium/total LDH activity) × 100%.

140

roo f

123

or P

Flow cytometry

USA). Cells treated with formononetin in the presence or absence of hypoxia treatment were allowed to swell by adding 100 ␮l of lysis buffer. The protein concentration was determined, and equal amount of protein and primary antibody were added to each well for 1 h in a 37◦ C incubator. Then the wells were washed 5 times and secondary antibody was added, the plate was then placed in a 22◦ C incubator for another 20 min. Stop buffer was added and the final value was determined at 450 nm using a spectrophotometer microplate reader.

uth

122

120

dA

121

(Sunrise®, Tecan, Sweden). Three control wells of medium and DMSO alone was used to provide the blanks for absorbance readings.

119

␣-secretase activity was measured following the manufacturer’s instructions (GBD, San Diego, CA,

3

165 166 167 168 169 170 171 172 173 174

175 176

177 178 179 180 181 182 183 184 185 186 187 188 189 190 191 192 193 194 195 196 197 198 199 200 201 202 203 204 205 206 207 208 209 210 211 212 213 214

220 221 222 223 224 225 226 227 228

229

230 231 232 233 234 235 236 237 238 239 240

241

Measurement of caspase-3 activity Caspase activity was assayed using caspase-3 activity assay kit (MBL) according to the manufacturer’s instructions. Briefly, cells were collected and washed with PBS, then resuspended in a cell lysis buffer. After incubation on ice for 10 min, the lysates were centrifuged for 20 min at 12,000 × g, and the supernatants were collected and protein concentrations were determined. Cell lysates (100 ␮g) were mixed with reaction buffer containing the DEVD-pNA substrate (200 ␮M) for caspase-3 activity. The absorbance was measured in the wells at 460 nm using an ELISA reader. RT-PCR measurement

256

siRNA-mediated knockdown

244 245 246 247 248 249 250 251 252 253 254

257 258 259 260

Un

243

co

255

To detect mRNA levels, total RNA was extracted with TRIZOL (Invitrogen), and converted to cDNA by reverse kit (Takara) according to the manufacturer’s instructions. ADAM10 sense primer, 5 CC ATgCTCATggAAgACAgTT-3 ; ADAM10 antisense primer, 5 -CCTTCTTCSCCSTSSSTSTgTCCA-3 ; ␤actin sense primer, 5 -TGTACGCCTCTGGCCG TACC-3 ; and ␤-actin antisense primer, 5 -CCACG TCACACTTCATGATGG-3 , PCR reactions were performed at 94◦ C for 30 s, 60◦ C for 1 min, and 68◦ C for 2 min during 40 cycles, followed by a final extension of 7 min at 68◦ C. The resulting PCR products were analyzed on a 1% agarose gel stained with ethidium bromide.

242

roo f

219

Statistical analysis

or P

218

ference MAX reagent (Invitrogen) according to the manufacturer’s instructions. The target sequences of the siRNAs specific for mouse ADAM10, or scrambled siRNA (Genechem, Shanghai, China) were as follows: ADAM10, 5 -GCAAAGATGATTGCTGCTTCG-3 ; Scramble 5 -GGTATATGCGCCATACACTACCC-3 . At 24 h post-siRNA treatment, cells were collected for different measurements.

All data in the text and figures are expressed as mean ± S.D. of at least three independent experiments. A one-way analysis of variance (ANOVA) followed by Dunnett’s or Tukey-Kramer’s post hoc tests was performed to compare groups. Mean values were considered significantly different at *p < 0.05, **p < 0.01, or ***p < 0.001. RESULTS

dA

217

proteins were transferred onto PVDF membranes, subsequently immunostained with the following primary antibodies against ADAM9 (1 : 500, Sigma), ADAM10 (1 : 2000, Abcam), ADAM17 (1 : 500, Sigma), and GAPDH (1 : 5000, Calbiochem). Following incubation with the appropriate horseradish peroxidase-conjugated secondary antibody (1 : 2,000) for 1 h at ambient temperature, the immunoblots were developed using the ECL system. Quantitative densitometric analyses were performed on digitized images of immunoblots with Quantity One software (Bio-Rad, Hercules, CA, USA). Representative blots from at least three independent experiments were shown.

cte

216

rre

215

M. Sun et al. / Formononetin Increases sA␤PP␣ and ADAM10 Level

uth

4

The siRNA method was performed according to previous work [48]. N2a-A␤PP cells were reverse transfected with 20 nM small interfering RNA (siRNA) duplexes by the use of Lipofectamine RNA inter-

Formononetin protects cells against hypoxia-induced cytotoxicity Using a cell culture model with hypoxic treatment, we confirmed the impairments of hypoxia treatment to cell viability by MTT and flow cytometry assay as previously reported [26]. To examine the neuroprotective effects of formononetin against hypoxia-induced cell viability loss, N2a-A␤PP cells were treated with hypoxia in the presence and absence of formononetin at different doses (0.1, 1, 10, 20, 50, and 100 ␮M) for 18 h, and results showed formononetin markedly attenuated hypoxia-induced cytotoxicity in a concentration-dependent manner (Fig. 2A). As the 10 ␮M formononetin was the minimum effective concentration in our system, it was used in the following experiments. To determine whether this effect was time-dependent, N2a-A␤PP cells were exposed to hypoxia condition at different time points in the presence or absence of 10 ␮M formononetin and results showed formononetin significantly attenuated hypoxia-induced cytotoxicity after 18 h of hypoxia treatment with cell viability 25% higher in formononetin group than control group (Fig. 2B). Similar results were obtained by flow cytometry experiments: hypoxia treatment significantly reduced cell viability, while pretreatment with formononetin notably increased living cell number and reduced cell death rate (Fig. 2C). Using EGb, a generally recognized antioxidant which could protect cells from oxidative damage [27] as positive control, we found formononetin also

261 262 263 264 265 266 267 268

269

270 271 272 273 274 275 276

277

278 279

280 281 282 283 284 285 286 287 288 289 290 291 292 293 294 295 296 297 298 299 300 301 302 303 304 305 306 307

M. Sun et al. / Formononetin Increases sA␤PP␣ and ADAM10 Level

A

5

roo f or P

Cell viability

Cell viability (% of control)

C

Treatment

FRM concentration (µ M)

D

dA

Cell viability (% of control)

LDH release (% of control)

uth

B

Hypoxia time (hours)

Treatment

Treatment

co

rre

Caspase 3 activity (fold increase)

cte

E

308 309 310 311 312 313 314 315 316 317

Un

Fig. 2. Protective effect of formononetin (FRM). A) N2a-A␤PP cells treated with different concentrations of FRM were collected for MTT assay 18 h later. B) N2a-A␤PP cells were treated at different hypoxia time points (0, 4, 8, 12, 18, 24 h) with or without 10 ␮M FRM pre-treatment and then measured by MTT assay. C) N2a-A␤PP cells were pre-treated with FRM, EGb, or the same amount of DMSO for 4 h and then processed with hypoxia treatment for another 18 h. At the end, cells were collected for flow cytometry assay. D) N2a-A␤PP cells were pre-treated with FRM, EGb, or the same amount of DMSO for 4 h and then processed with hypoxia treatment for another 18 h. Cells lysates were collected for LDH assay, and (E) for caspase-3 activity assay. All data were represented as a mean ± S.D. from triplicate independent experiments. *p < 0.05, **p < 0.01, ***p < 0.001.

had a protective effect similar with that of EGb (Fig. 2C). Accordingly, we also evaluated LDH activity in culture medium as an indicator of cell damage and showed that 10 ␮M formononetin pretreatment reduced hypoxia-mediated LDH release by nearly 40%, which further confirmed its protective role (Fig. 2D). As caspase-3 activity is an indicator of apoptosis [28], we measured its level under hypoxia in the presence or absence of different chemicals. In agreement with a previous report [29], we also observed a

significant increase of caspase-3 activity after hypoxia treatment; meanwhile in formononetin pretreatment groups, we found that the casapse-3 activity showed a significant decrease compared with the levels of those without formononetin pretreatment (Fig. 2E). Formononetin could stimulate α-secretase activity Previous reports have shown that hypoxia treatment would induce a calcium influx, activate down-stream

318 319 320 321 322

323

324 325

333 334 335 336 337 338 339 340 341 342 343 344 345 346

roo f

332

or P

331

Formononetin increases α-secretase activity by upregulating ADAM10 protein level, but not ADAM 9 or ADAM17 As we have found formononetin treatment increases ␣-secretase activity, we further investigated its effect

dA

330

A

cte

329

level was significantly reduced while the total amount of A␤PP did not change (Fig. 3B). In addition to the changes in sA␤PP␣, we showed that formononetin could also upregulate CTF-␣ protein levels compared to the hypoxia treatment group (Fig. 3B, C). We next measured ␣-secretase activity and noticed that after treatment with formononetin, ␣-secretase activity showed a threefold increase compared to the hypoxia group (Fig. 4A). Combined with the elevated CTF-␣ protein level, our results above indicated that formononetin could shift A␤PP processing toward the non-amyloid pathway by increasing ␣-secretase activity.

1.5

1.0

0.5

0.0

rre

328

caspase-3 activity, and down-regulate sA␤PP␣ levels [30, 34]. Moreover, sA␤PP␣ is also known to be a potent mediator that attenuates excessive calcium entry and excitotoxicity [32, 33]; therefore, formononetin might protect cells from hypoxia-induced toxicity by upregulating sA␤PP␣. Indeed, we found formononetin could upregulate sA␤PP␣ in a dose-dependent manner under normoxia condition (data not shown). So we continued to examine the effect of formononetin on sA␤PP␣ levels under hypoxia treatment and found, as previously reported, that sA␤PP␣ was significantly decreased under hypoxic conditions [34], while formononetin pretreatment significantly increased sA␤PP␣ secretion (Fig. 3A). The results above suggest that formononetin may exert its protective effect by increasing sA␤PP␣ secretion under hypoxic conditions. Furthermore, we found this increase in sA␤PP␣ was conducted by shifting A␤PP processing to the nonamyloid cleavage pathway. As shown in Fig. 3B, in agreement with previous reports [31, 45, 58], we also found that after hypoxia treatment the CTF-␣ protein

IP

WB

B

1.25 1.00 0.75

co

327

Un

326

M. Sun et al. / Formononetin Increases sA␤PP␣ and ADAM10 Level

uth

6

C

1.25 1.00 0.75

0.50

0.50

0.25

0.25

0.00

0.00

Fig. 3. A␤PP processing was measured after formononetin (FRM) administration. N2a-A␤PP cells were pre-treated with FRM, EGb, or same amount of DMSO for 4 h and then processed with hypoxia treatment for another 18 h. At the end, cells lysates were collected for western blot or immuno-precipitation to detect (A) sA␤PP␣ secretion level; (B) full length A␤PP protein level; and (C) CTF-␣ protein level. All data were represented as a mean ± S.D. from triplicate independent experiments. *p < 0.05, **p < 0.01, ***p < 0.001.

347 348 349 350 351 352 353 354 355 356 357 358 359

360 361 362

363 364

7

Un

co

rre

cte

dA

uth

or P

roo f

M. Sun et al. / Formononetin Increases sA␤PP␣ and ADAM10 Level

Fig. 4. Formononetin (FRM) could increase ␣-secretase by upregulating ADAM10 mRNA level. N2a-A␤PP cells were pre-treated with or without FRM for 4 h and then hypoxia treatment for an additional 18 h. After that, cells lysates were collected for different measurements as follows: (A) ␣-secretase activity; (B) pre-mature form of ADAM10 protein level; (C) mature form of ADAM10 protein level. D) The ratio of mature form of ADAM10 versus pre-mature form of ADAM10; (E) ADAM 9 protein level, and (F) ADAM17 protein level. Total mRNA was also isolated for detecting (G) ADAM10 mRNA level. All data were represented as a mean ± S.D. from triplicate independent experiments. *p < 0.05, **p < 0.01, ***p < 0.001.

375 376 377 378 379 380 381 382 383 384 385 386 387

roo f

374

or P

373

1.25 1.00 0.75 0.50 0.25 0.00

2.5 2.0 1.5 1.0 0.5 0.0

2.5

ADAM10 mRNA level (% of control)

372

dA

371

cte

370

Since we found that formononetin could upregulate ADAM10 under hypoxic condition, followed by increasing sA␤PP␣ secretion levels, we next wondered whether formononetin’s effect on ADAM10 expression was hypoxia dependent. As shown in Fig. 5, we found under normoxia condition, when N2a-A␤PP cells were treated with formononetin, the total protein level of ADAM10 and its mRNA level were both increased (Fig. 5C, D, lane 1 compared with lane 2). Furthermore, in addition to increasing ADMA10, formononetin administration could also upregulate ␣secretase activity (Fig. 5B, lane 1 and lane 2), which indicated that formononetin could increase sA␤PP␣ by elevating ␣-secretase activity and ADAM10 transcriptional levels independent of hypoxia. At last, we wanted to confirm whether the cell protective effect under hypoxic conditions was conducted by the ADAM10-sA␤PP␣ pathway activated by formononetin treatment. For this purpose, we used the ␣-secretase specific inhibitor, TAPI-2, which could

rre

369

Formononetin’s protective effect is mainly dependent on the sAβPPα pathway

co

368

Un

367

on the components of ␣-secretase complex to clarify the underlying mechanism. As shown in Fig. 4B and C, we found that the levels of both pre-mature and mature ADAM10 were decreased under hypoxia treatment and that formononetin treatment could significantly attenuate this decrease, as well as increase the mature/immature ratio of it compared with the hypoxia group (Fig. 4D). Meanwhile, other potential ␣-secretase candidates, ADAM9 and ADAM17, did not exhibit a statistically significant change under different experimental treatments (Fig. 4E, F). Since our results showed that not only the mature form of ADAM10 was increased by formononetin treatment but also the pre-mature form, a question was raised regarding whether or not formononetin treatment could increase ADAM10 at the transcriptional level. As shown in Fig. 4G, employing conventional PCR, we found that the mRNA level of ADAM10 was significantly higher in the formononetin treatment group compared to the hypoxia group, suggesting that under hypoxic condition, formononetin treatment could recover ADAM10 at the transcriptional level.

sAβPPα protein level (% of control)

366

α-secretase activity (Fold Increace)

365

M. Sun et al. / Formononetin Increases sA␤PP␣ and ADAM10 Level

uth

8

2.0 1.5 1.0 0.5 0.0

Fig. 5. sA␤PP␣ secretion and ␣-secretase activity were totally abolished by TAPI-2. A) N2a-A␤PP cells were treated with formononetin (FRM) in the presence or absence of TAPI-2 for 24 h and then cells lysates were collected for different measurements as follows: (A) sA␤PP␣ protein level; (B) ␣-secretase activity; and (C) ADAM10 protein level. Total mRNA was also isolated for detecting (D) ADAM10 mRNA level. All data were represented as a mean ± S.D. from triplicate independent experiments. *p < 0.05, **p < 0.01, ***p < 0.001.

388 389

390 391 392 393 394 395 396 397 398 399 400 401 402 403 404 405 406 407 408 409

M. Sun et al. / Formononetin Increases sA␤PP␣ and ADAM10 Level

418 419 420

roo f

417

or P

416

uth

415

dA

414

cte

413

ment (Fig. 6C). Meanwhile both MTT and flow cytometry assays showed a decrease of cell viability in formononetin with the TAPI-2 group compared with the formononetin alone group under hypoxic conditions (Fig. 6D–I). Furthermore, ADAM10 siRNA treatment also appeared to give a similar result. As shown in Fig. 7D–J, flow cytometry assay showed a nearly 40% decrease of living cell number in the hypoxia group compared with control, and formononetin treatment could attenuate hypoxia-induced cell impairment, while ADAM10 siRNA could almost

rre

412

significantly inhibit ␣-secretase activity and further decrease sA␤PP␣ production (Fig. 5A, B) without influencing ADAM10 expression (Fig. 5C, D), and ADAM10 siRNA which could inhibit sA␤PP␣ secretion while diminish ADAM10 expression (Fig. 7A, B, C). Treating the cells with TAPI-2 and formononetin for a total of 6 h followed by an additional 18 h of hypoxia treatment, we found while the sA␤PP␣ secretion and ␣-secretase activity were almost totally abolished (Fig. 6A, B), the mRNA level of ADAM10 was hardly influenced by TAPI-2 under hypoxia treat-

co

411

Un

410

9

Fig. 6. The neuroprotective effect of formononetin (FRM) was inhibited by TAPI-2. N2a-A␤PP cells were administrated with hypoxia treatment in the presence or absence of pre-treatment of FRM and TAPI-2. 18 h later, cells lysates or total mRNA was collected for different measurements as follows: (A) sA␤PP␣ protein level; (B) ␣-secretase activity; (C) ADAM10 mRNA level. After 18 h hypoxia treatment, cells were also collected for detecting apoptosis rate or cell viability. D–H) Flow cytometry assay for neuro-apoptosis rate measurement in the presence or absence of FRM or TAPI-2. I) Viability of N2a-A␤PP cell after 18 h hypoxia treatment with FRM and TAPI-2 was measured by MTT assay. All data were represented as a mean ± S.D. from triplicate independent experiments. *p < 0.05, **p < 0.01, ***p < 0.001.

421 422 423 424 425 426 427 428 429 430 431

M. Sun et al. / Formononetin Increases sA␤PP␣ and ADAM10 Level

Un

co

rre

cte

dA

uth

or P

roo f

10

Fig. 7. The neuroprotective effect of formononetin (FRM) was inhibited by ADAM10 siRNA. N2a-A␤PP cells were transfected by ADAM10 siRNA for 24 h, and then (A) ADAM10 protein level; (B) ␣-secretase activity; and (C) ADAM10 mRNA level was detected. Cells were exposed to hypoxia in the presence or absence of FRM pre-treatment for 18 h, and then cells were collected for different measurements as follows: D–H) flow cytometry assay for neuro-apoptosis rate measurement in the presence or absence of FRM and ADAM10 siRNA. I) MTT assay for cell viability measurement after 18 h hypoxia treatment in the presence or absence of FRM and ADAM10 siRNA. J) Caspase-3 activity after 18 h hypoxia treatment with FRM and ADAM10 siRNA was measured. All data were represented as a mean ± S.D. from triplicate independent experiments. *p < 0.05, **p < 0.01, ***p < 0.001.

432 433 434 435 436

abolish formononetin -induced protective effects and reduction in caspase-3 activity. MTT assay also showed a similar result (Fig. 7I). All results above indicated that the protective effect of formononetin against hypoxia was mainly conducted by the ADAM10sA␤PP␣ pathway.

DISCUSSION In the A␤PP non-amyloid cleavage pathway, A␤PP would be cleaved by ␣-secretase and thus produce the neuroprotective fragment sA␤PP␣ [35, 43]. Induced sA␤PP␣ secretion might have additional advan-

437

438 439 440

M. Sun et al. / Formononetin Increases sA␤PP␣ and ADAM10 Level

449 450 451 452 453 454 455 456 457 458 459 460 461 462 463 464 465 466 467 468 469 470 471 472 473 474 475 476 477 478 479 480 481 482 483 484 485 486 487 488 489 490 491 492

roo f

448

or P

447

uth

446

dA

445

nificantly affected by ischemic insult [55], while another study showed that BACE1 levels were unchanged in human neuroblastoma SH-SY5Y cells subjected to chronic hypoxic treatments [34]. Recently, researchers have found both BACE-1 [31, 53] and APH-1 [45, 46] are upregulated by elevated HIF1␣ after hypoxia treatment, and the amount of C99 as well as the ratio of C99/C83 would also be increased. In agreement with the latter two studies, we also found a significant increase in BACE activity and CTF-␤ level (supplementary Figure 1; available online: http://www.j-alz.com/issues/28/vol284.html#supplementarydata03) as well as a decrease of CTF-␣. Enzymes acting as ␣-secretase complex in the A␤PP non-amyloid pathway have been identified as members of ADAM family. It has also been found that under hypoxic condition ␣-secretase activity and sA␤PP␣ would decrease significantly [45, 56, 57]. Three of these membrane-anchored zinc-dependent metalloproteinases, ADAM10 as well as ADAM17 and ADAM9, are the candidate enzymes that show ␣-secretase activity [47]. Among them, although ADAM10 has been extensively researched, the results are still controversial. Some studies found ADAM10 and TACE level significantly decreased after hypoxia treatment [34, 58], yet other studies, using primary cell of AD transgenic mice overexpressing mutant A␤PP or cerebral microvascular smooth muscle cells as a model, found that ADAM10 was markedly increased in the early stage of ischemic insult or hypoxia treatment and afterwards decreased [55, 57]. Furthermore, another study found in the SH-SY5Y cell line that hypoxia treatment would change APH-1 and BACE-1 levels but not ADAM10 or TACE [49]. While it is hard to reconcile these results, differing experimental procedures, such as cells/tissues examined, culture conditions, the percentage of O2 utilized, or duration of hypoxic treatment, could all contribute to discrepant results. In agreement with some previous works, we found a significant decrease in ADAM10 protein after 18 h of hypoxic treatment coincidently with a decrease in sA␤PP␣ secretion and CTF-␣ level. SIRT1 is one of the seven mammalian proteins of the sirtuin family of NAD (+)-dependent deacetylases, and its hyperactivity might be able to prevent AD pathology both in vitro and in vivo. It has been recently found that SIRT1 could directly activate transcription of ADAM10, elevating both mature and immature forms of ADAM10, but not ADAM9 or ADAM17, and further increasing the activity of ␣secretase [50]. Additionally, there was a study showing

cte

444

rre

443

tages, for various studies have strongly established that secreted sA␤PP␣ possesses potent neurotrophic and neuroprotective activities against excitotoxic and oxidative assaults [32, 36], JNK-mediated apoptosis [37], and the pro-apoptotic action of mutant presenlin1 by activating the transcription factor NF-␬B [38]. Moreover, sA␤PP␣ stimulates neurite outgrowth [39], regulates synaptogenesis [40], has trophic effects on cerebral neurons in culture [41], stabilizes neuronal calcium homeostasis, and protects hippocampal and cortical neurons against the toxic effects of glutamate and A␤ peptides [42]. Also, it has been shown that intracerebroventricular administration of secreted forms of sA␤PP␣ to amnestic mice had potent memory-enhancing effects and blocked learning deficits induced by scopolamine [18]. Therefore, increased production of sA␤PP␣ protein by upregulating ADAMs would potentially be beneficial for AD [44, 50]. Cerebral hypoxia results from an insufficient oxygen supply to the brain and causes neuronal damage in vulnerable brain areas [60]. Accumulating evidence indicates that cerebral hypoxia/stroke significantly increases AD risk [61–63]. Multiple studies have confirmed that hypoxia treatment could cause cell damage [review see 68], one of the mechanisms being the activation of caspase-3. Caspase-3 is the most abundant of the known caspases in neural cells, and appears to play a crucial role both during normal development and in situations of hypoxic injury [64]. It has been found that under hypoxic conditions, caspase-3 activity is stimulated by increased calcium influx, which is the common event after exposure to hypoxia [65, 66]. As sA␤PP␣ is also known to be a potent mediator attenuating excessive calcium entry and excitotoxicity [32, 67], we found that formononetin’s inhibition effects on caspase-3 activity was mainly conducted by increasing sA␤PP␣ secretion level. Administration of ADAM10 siRNA, which would further inhibit sA␤PP␣ secretion, could largely abolished formononetin’s inhibition effect on caspase-3 activity and its neuroprotective effect against hypoxia. Although hypoxia/ischemia is known to be a risk factor for AD, the detailed mechanism is still unclear, and there are conflicting results concerning the relationship between hypoxia and A␤PP processing pathways. Some studies [54, 59] found that both activity and expression of BACE1 were significantly increased in rats under transient cerebral ischemia. Another study using AD transgenic mice overexpressing mutant A␤PP, however, showed that neither expression nor activity of BACE was sig-

co

442

Un

441

11

493 494 495 496 497 498 499 500 501 502 503 504 505 506 507 508 509 510 511 512 513 514 515 516 517 518 519 520 521 522 523 524 525 526 527 528 529 530 531 532 533 534 535 536 537 538 539 540 541 542 543 544

552 553 554 555 556 557 558 559 560 561 562 563 564 565 566 567 568 569 570 571 572 573 574

575

576 577 578 579 580 581 582 583 584 585 586 587

588

589 590 591 592

[4]

[5]

[6]

ACKNOWLEDGMENTS

We thank Dr. Sangram S. Sisodia (University of Chicago, Chicago) for providing Mouse N2a neuroblastoma cells stably expressing A␤PPsw. This work was supported by the National Natural Science Foundation of China (NSFC; Grants No. 30670414, No.30830120 No.30973145 and No.81171015), Doctoral Fund of Ministry of Education (20090001 110058).and the National High Technology Research and Development Program of China (973 Program No. 2012CB911004). Authors’ disclosures available online (http://www.jalz.com/disclosures/view.php?id=1037). REFERENCES [1]

Kulling SE, Lehmann L, Metzler M (2002) Oxidative metabolism and genotoxic potential of major isoflavone phytoestrogens. J Chromatogr B Analyt Technol Biomed Life Sci 777, 211-218.

roo f

551

[3]

Huh JE, Nam DW, Baek YH, Kang JW, Park DS, Choi DY, Lee JD (2011) Formononetin accelerates wound repair by the regulation of early growth response factor-1 transcription factor through the phosphorylation of the ERK and p38 MAPK pathways. Int Immunopharmacol 11, 46-54. Chen HQ, Wang XJ, Jin ZY, Xu XM, Zhao JW, Xie ZJ (2008) Protective effect of isoflavones from trifolium pratense on dopaminergic neurons. Neurosci Res 62, 123-130. Occhiuto F, Zangla G, Samperi S, Palumbo DR, Pino A, De Pasquale R, Circosta C (2008) The phytoestrogenic isoflavones from Trifolium pratense L. (Red clover) protects human cortical neurons from glutamate toxicity. Phytomedicine 15, 676-682. Occhiuto F, Palumbo DR, Samperi S, Zangla G, Pino A, De Pasquale R, Circosta C (2009) The isoflavones mixture from Trifolium pratense L. protects HCN 1-A neurons from oxidative stress. Phytother Res 23, 192-196. Yu DH, Bao YM, An LJ, Yang M (2009) Protection of PC12 cells against superoxide-induced damage by isoflavonoids from astragalus mongholicus. Biomed Environ Sci 22, 50-54. Yu D, Duan Y, Bao Y, Wei C, An L (2005) Isoflavonoids from Astragalus mongholicus protect PC12 cells from toxicity induced by L-glutamate. J Ethnopharmacol 98, 89-94. Anderson JJ, Holtz G, Baskin PP, Wang R, Mazzarelli L, Wagner SL, Menzaghi F (1999) Reduced cerebrospinal fluid levels of alpha-secretase-cleaved amyloid precursor protein in aged rats: Correlationwith spatialmemory deficits. Neuroscience 93, 1409-1420. Kern A, Roempp B, Prager K, Walter J, Behl C (2006) Down-regulation of endogenous amyloid precursor protein processing due to cellular aging. J Biol Chem 281, 2405-2413. Jendroska K, Hoffmann OM, Patt S (1997) Amyloid beta peptide and precursor protein (ABPP) in mild and severe brain ischemia. Ann N Y Acad Sci 826, 401-405. Koistinaho J, Pyykonen I, Keinanen R, Hokfelt T (1996) Expression of beta-amyloid precursor protein mRNAs following transient focal ischaemia. NeuroReport 7, 27272731. Pietrzik CU, Hoffmann J, St¨ober K, Chen CY, Bauer C, Otero DA, Roch JM, Herzog V (1998) From differentiation to proliferation: The secretory amyloid precursor protein as a local mediator of growth in thyroid epithelial cells. Proc Natl Acad Sci U S A 95, 1770-1775. Caill´e I, Allinquant B, Dupont E, Bouillot C, Langer A, M¨uller U, Prochiantz A (2004) Soluble form of amyloid precursor protein regulates proliferation of progenitors in the adult subventricular zone. Development 131, 2173-2181. Gralle M, Ferreira ST (2007) Structure and functions of the human amyloid precursor protein: The whole is more than the sum of its parts. Prog Neurobiol 82, 11-32. Chen CW, Boiteau RM, Lai WF, Barger SW, Cataldo AM (2006) sA␤PPalpha enhances the transdifferentiation of adult bone marrow progenitor cells to neuronal phenotypes. Curr Alzheimer Res 3, 63-70. Chasseigneaux S, Dinc L, Rose C, Chabret C, Coulpier F, Topilko P, Mauger G, Allinquant B (2011) Secreted amyloid precursor protein ␤ and secreted amyloid precursor protein ␣ induce axon outgrowth in vitro through Egr1 signaling pathway. PLoS One 27, e16301. Furukawa K, Sopher BL, Rydel RE, Begley JG, Pham DG, Martin GM, Fox M, Mattson MP (1996) Increased activityregulating and neuroprotective efficacy of alpha-secretasederived secreted amyloid precursor protein conferred by a C-terminal heparin-binding domain. J Neurochem 67, 18821896.

or P

550

[2]

[7]

[8]

dA

549

cte

548

rre

547

that, as one of the chemicals belonging to isoflavone, formononetin could increase both the activity and expression of SIRT1 [51]. Combined with the aforementioned results, the fact that formononetin could increase SIRT1 expression and that SIRT1 had no effects on ADAM9 or ADAM17 gives us a clue that formononetin probably regulates ADAM10 by modulating SIRT1 level. Here we report that after formononetin administration, the ␣-secretase activity of N2a-A␤PP cells was significantly higher than the hypoxia treatment group. Our results also show both that mature and immature forms of ADAM10 were elevated while ADAM9 and ADAM17 did not change. Interestingly, we also found that the ratio of ADAM10 mature to immature form was increased by formononetin treatment, which indicates that formononetin may not only regulate ADAM10 on a transcriptional level, but also influence ADAM10 protein trafficking and subcellular location. In summary, we demonstrated for the first time that formononetin prevented hypoxia-induced neurotoxicity by increasing sA␤PP␣ expression in N2a-A␤PP cells, and we further found that the effect of formononetin on sA␤PP␣ level was mainly mediated by upregulating ADAM10 at the transcriptional level and further increasing ␣-secretase activity. These findings provide novel insights on formononetin’s neuroprotective mechanisms and the signaling pathways involved, and could be valuable to future therapeutic research.

co

546

Un

545

M. Sun et al. / Formononetin Increases sA␤PP␣ and ADAM10 Level

uth

12

[9]

[10]

[11]

[12]

[13]

[14]

[15]

[16]

[17]

593 594 595 596 597 598 599 600 601 602 603 604 605 606 607 608 609 610 611 612 613 614 615 616 617 618 619 620 621 622 623 624 625 626 627 628 629 630 631 632 633 634 635 636 637 638 639 640 641 642 643 644 645 646 647 648 649 650 651 652 653 654 655 656 657

M. Sun et al. / Formononetin Increases sA␤PP␣ and ADAM10 Level

665 666

[20]

667 668 669

[21]

670 671 672

[22]

673 674 675

[23]

676 677 678 679

[24]

680 681 682 683 684 685

[25]

686 687 688 689

[26]

690 691 692 693

[27]

694 695 696 697 698 699

[28]

700 701 702

[29]

703 704 705 706

[30]

707 708 709

[31]

710 711 712 713

[32]

714 715 716 717

[33]

718 719 720 721 722

[34]

[37]

[38]

[39]

[40]

roo f

664

or P

663

[36]

uth

[19]

ADAM10 and TACE, without altering their mRNA levels. Brain Res 1099, 18-24. Selkoe DJ (2001) Alzheimer’s disease: Genes, proteins, and therapy. Physiol Rev 81, 741-766. Clement AB, Gimpl G, Behl C (2010) Oxidative stress resistance in hippocampal cells is associated with altered membrane fluidity and enhanced nonamyloidogenic cleavage of endogenous amyloid precursor protein. Free Radic Biol Med 48, 1236-1241. K¨ogel D, Schomburg R, Copanaki E, Prehn JH (2005) Regulation of gene expression by the amyloid precursor protein: Inhibition of the JNK/c-Jun pathway. Cell Death Differ 12, 1-9. Guo Q, Robinson N, Mattson MP (1998) Secreted betaamyloid precursor protein counteracts the proapoptotic action of mutant presenilin-1 by activation of NF-kappaB and stabilization of calcium homeostasis. J Biol Chem 273, 1234112351. Small DH, Nurcombe V, Reed G, Clarris H, Moir R, Beyreuther K, Masters CL (1994) A heparin-binding domain in the amyloid protein precursor of Alzheimer’s disease is involved in the regulation of neurite outgrowth. J Neurosci 14, 2117-2127. Morimoto T, Ohsawa I, Takamura C, Ishiguro M, Kohsaka S (1998) Involvement of amyloid precursor protein in functional synapse formation in cultured hippocampal neurons. J Neurosci Res 51, 185-195. Araki W, Kitaguchi N, Tokushima Y, Ishii K, Aratake H, Shimohama S, Nakamura S, Kimura J (1991) Trophic effect of beta-amyloid precursor protein on cerebral cortical neurons in culture. Biochem Biophys Res Commun 181, 265-271. Furukawa K, Barger SW, Blalock EM, Mattson MP (1996) Activation of K+ channels and suppression of neuronal activity by secreted beta-amyloid-precursor protein. Nature 379, 74-78. Hou Y, Yu YB, Liu G, Luo Y (2010) A natural squamosamide derivative FLZ reduces amyloid-beta production by increasing non-amyloidogenic AbetaPP processing. J Alzheimers Dis 18, 153-165. Schroeder A, Fahrenholz F, Schmitt U (2010) Effect of a dominant-negative form of ADAM10 in a mouse model of Alzheimer’s disease. J Alzheimers Dis 16, 309-314. Wang R, Zhang YW, Zhang X, Liu R, Zhang X, Hong S, Xia K, Xia J, Zhang Z, Xu H (2006) Transcriptional regulation of APH-1A and increased gamma-secretase cleavage of APP and Notch by HIF-1 and hypoxia. FASEB J 20, 1275-1277. Li L, Zhang X, Yang D, Luo G, Chen S, Le W (2009) Hypoxia increases Abeta generation by altering beta- and gammacleavage of APP. Neurobiol Aging 30, 1091-1098. Endres K, Fahrenholz F (2010) Upregulation of the alphasecretase ADAM10 – risk or reason for hope? FEBS J 277, 1585-1596. Cagavi Bozkulak E, Weinmaster G (2009) Selective use of ADAM10 and ADAM17 in activation of Notch1 signaling. Mol Cell Biol 21, 5679-5695. Li QY, Wang HM, Wang ZQ, Ma JF, Ding JQ, Chen SD (2010) Salidroside attenuates hypoxia-induced abnormal processing of amyloid precursor protein by decreasing BACE1 expression in SH-SY5Y cells. Neurosci Lett 481, 154-158. Donmez G, Wang D, Cohen DE, Guarente L (2010) SIRT1 suppresses beta-amyloid production by activating the alphasecretase gene ADAM10. Cell 142, 320-332. Rasbach KA, Schnellmann RG (2008) Isoflavones promote mitochondrial biogenesis. J Pharmacol Exp Ther 325, 536543.

dA

662

[35]

[41]

[42]

cte

661

rre

660

Meziane H, Dodart JC, Mathis C, Little S, Clemens J, Paul SM, Ungerer A (1998) Memory-enhancing effects of secreted forms of the beta-amyloid precursor protein in normal and amnestic mice. Proc Natl Acad Sci U S A 95, 12683-12688. Thornton E, Vink R, Blumbergs PC, Van Den Heuvel C (2006) Soluble amyloid precursor protein alpha reduces neuronal injury and improves functional outcome following diffuse traumatic brain injury in rats. Brain Res 1094, 38-46. Bart De Strooper, Robert Vassar, Todd Golde (2010) The secretases: Enzymes with therapeutic potential in Alzheimer disease. Nat Rev Neurol 6, 99-107. Yu AC, Lau LT (2000) Expression of interleukin-1 alpha, tumor necrosis factor alpha and interleukin-6 genes in astrocytes under ischemic injury. Neurochem Int 36, 369-377. Yu J, Sun M, Chen Z, Lu J, Liu Y, Zhou L, Xu X, Fan D, Chui D (2010) Magnesium modulates amyloid-beta protein precursor trafficking and processing. J Alzheimers Dis 20, 1091-1106. Mi Li, Miao Sun, Yi Liu, Jia Yu, Huan Yang, Dongsheng Fan, Dehua Chui (2010) Copper down-regulates neprilysin activity through modulation of neprilysin degradation. J Alzheimers Dis 19, 161-169. Wiltfang J, Esselmann H, Cupers P, Neumann M, Kretzschmar H, Beyermann M, Schleuder D, Jahn H, Ruther E, Kornhuber J, Annaert W, De SB, Saftig P (2001) Elevation of beta-amyloid peptide 2-42 in sporadic and familial Alzheimer’s disease and its generation in PS1 knockout cells. J Biol Chem 276, 42645-42657. Zhao G, Mao G, Tan J, Dong Y, Cui MZ, Kim SH, Xu X (2004) Identification of a new presenilin-dependent zeta-cleavage site within the transmembrane domain of amyloid precursor protein. J Biol Chem 279, 50647-50650. Tulsawani R, Kelly LS, Fatma N, Chhunchha B, Kubo E, Kumar A, Singh DP (2010) Neuroprotective effect of peroxiredoxin 6 against hypoxia-induced retinal ganglion cell damage. BMC Neurosc 11, 125. Yuan Luo, Julie V, Smith, Vijaykumar Paramasivam, Adam Burdick, Kenneth J, Curry, Justin P, Buford, Ikhlas Khan, William J, Netzer, Huaxi Xu, Peter, Butko (2002) Inhibition of amyloid-␤ aggregation and caspase-3 activation by the Ginkgo biloba extract EGb761. Proc Natl Acad Sci U S A 99, 12197-12202. Harada J, Sugimoto M (1999) Activation of caspase-3 in betaamyloid-induced apoptosis of cultured rat cortical neurons. Brain Res 842, 311-323. Delivoria-Papadopoulos M, Mishra OP (2007) Mechanism of activation of caspase-9 and caspase-3 during hypoxia in the cerebral cortex of newborn piglets: The role of nuclear Ca2+ -influx. Neurochem Re 32, 401-405. Bickler PE, Fahlman CS, Ferriero DM (2004) Hypoxia increases calcium flux through cortical neuron glutamate receptors via protein kinase C. J Neurochem 88, 878-884. Zhang X, Zhou K, Wang R, Cui J, Lipton SA, Liao FF, Xu H, Zhang YW (2007) Hypoxia-inducible factor 1alpha (HIF1alpha)-mediated hypoxia increases BACE1 expression and beta-amyloid generation. J Biol Chem 282, 10873-10880. Mattson MP, Cheng B, Culwell AR, Esch FS, Lieberburg I, Rydel RE (1993) Evidence for excitoprotective and intraneuronal calcium-regulating roles for secreted forms of the beta-amyloid precursor protein. Neuron 10, 243-254. Mattson MP (1997) Cellular actions of beta-amyloid precursor protein and its soluble and fibrillogenic derivatives. Physiol Rev 77, 1081-1132. Amy J Marshall, Marcus Rattray, Peter FT Vaughan (2006) Chronic hypoxia in the human neuroblastoma SH-SY5Y causes reduced expression of the putative ␣-secretases,

co

[18]

659

Un

658

[43]

[44]

[45]

[46]

[47]

[48]

[49]

[50]

[51]

13

723 724 725 726 727 728 729 730 731 732 733 734 735 736 737 738 739 740 741 742 743 744 745 746 747 748 749 750 751 752 753 754 755 756 757 758 759 760 761 762 763 764 765 766 767 768 769 770 771 772 773 774 775 776 777 778 779 780 781 782 783 784 785 786 787

[53]

795 796 797 798 799

[54]

800 801 802

[55]

803 804 805

[56]

806 807 808 809

[57]

810 811 812 813 814

[58]

815 816 817 818 819

[59]

[62]

[63] [64]

[65]

roo f

794

or P

793

[61]

Koistinaho M, Koistinaho J (2005) Interactions between Alzheimer’s disease and cerebral ischemia–focus on inflammation Brain Res. Brain Res Rev 48, 240-250. Tatemichi TK, Paik M, Bagiella E, Desmond DW, Stern Y, Sano M, Hauser WA, Mayeux R (1994) Risk of dementia after stroke in a hospitalized cohort: Results of a longitudinal study. Neurology 44, 1885-1189. Kokmen E, Whisnant JP, O’Fallon WM, Chu CP, Beard CM (1996) Dementia after ischemic stroke: A population-based study in Rochester, Minnesota (1960–1984). Neurology 46, 154-159. Kalaria RN (2000) The role of cerebral ischemia in Alzheimer’s disease. Neurobiol Aging 21, 321-330. Wang X, Karlsson JO, Zhu C, Bahr BA, Hagberg H, Blomgren K (2001) Caspase-3 activation after neonatal rat cerebral hypoxia-ischemia. Biol Neonate 79, 172-179. Delivoria-Papadopoulos M, Mishra OP (2007) Mechanism of activation of caspase-9 and caspase-3 during hypoxia in the cerebral cortex of newborn piglets: The role of nuclear Ca2+-influx. Neurochem Res 32, 401-405. Guo Z, Shi F, Zhang L, Zhang H, Yang J, Li B, Jia J, Wang X (2010) Critical role of L-type voltage-dependent Ca2+ channels in neural progenitor cell proliferation induced by hypoxia. Neurosci Lett 478, 156-160. Mattson MP (1997) Cellular actions of beta-amyloid precursor protein and its soluble and fibrillogenic derivatives. Physiol Rev 77, 1081-1132. Banasiak KJ, Xia Y, Haddad GG (2000) Mechanisms underlying hypoxia-induced neuronal apoptosis. Prog Neurobiol 62, 215-249.

uth

792

[60]

[66]

[67]

dA

791

cte

790

Kojro E, F¨uger P, Prinzen C, Kanarek AM, Rat D, Endres K, Fahrenholz F, Postina R (2010) Statins and the squalene synthase inhibitor zaragozic acid stimulate the non-amyloidogenic pathway of amyloid-beta protein precursor processing by suppression of cholesterol synthesis. J Alzheimers Dis 20, 1215-1231. Guglielmotto M, Aragno M, Autelli R, Giliberto L, Novo E, Colombatto S, Danni O, Parola M, Smith MA, Perry G, Tamagno E, Tabaton M (2009) The up-regulation of BACE1 mediated by hypoxia and ischemic injury: Role of oxidative stress and HIF1alpha. J Neurochem 108, 1045-1056. Wen Y, Onyewuchi O, Yang S, Liu R, Simpkins JW (2004) Increased beta-secretase activity and expression in rats following transient cerebral ischemia. Brain Res 1009, 1-8. Lee PH, Hwang EM, Hong HS, Boo JH, Mook-Jung I, Huh K (2006) Effect of ischemic neuronal insults on amyloid precursor protein processing. Neurochem Res 31, 821-827. Webster NJ, Green KN, Peers C, Vaughan PF (2002) Altered processing of amyloid precursor protein in the human neuroblastoma SH-SY5Y by chronic hypoxia. J Neurochem 83, 1262-1271. Webster NJ, Green KN, Settle VJ, Peers C, Vaughan PF (2004) Altered processing of the amyloid precursor protein and decreased expression of ADAM 10 by chronic hypoxia in SH-SY5Y: No role for the stress-activated JNK and p38 signalling pathways. Mol Brain Res 130, 161-169. Auerbach ID, Vinters HV (2006) Effects of anoxia and hypoxia on amyloid precursor protein processing in cerebral microvascular smooth muscle cells. J Neuropathol Exp Neurol 65, 610-620. Xue S, Jia L, Jia J (2006) Hypoxia and reoxygenation increased BACE1 mRNA and protein levels in human neuroblastoma SH-SY5Y cells. Neurosci Lett 405, 231-235.

rre

[52]

789

co

788

M. Sun et al. / Formononetin Increases sA␤PP␣ and ADAM10 Level

Un

14

[68]

820 821 822 823 824 825 826 827 828 829 830 831 832 833 834 835 836 837 838 839 840 841 842 843 844 845 846 847 848 849