Differential expression of neuroblastoma cellular proteome due to AICD overexpression

Share Embed


Descripción

1

Journal of Alzheimer’s Disease xx (20xx) x–xx DOI 10.3233/JAD-130695 IOS Press

3

roo f

2

Differential Expression of Neuroblastoma Cellular Proteome due to AICD Overexpression

or P

1

Arunabha Chakrabarti1 , Kasturi Roy1 and Debashis Mukhopadhyay∗

5

Structural Genomics Division, Saha Institute of Nuclear Physics, Kolkata, WB, India

6

Handling Associate Editor: Inga Zerr

uth

4

Accepted 10 August 2013

16

Keywords: Alzheimer’s disease, A␤PP intracellular domain, 2D-DIGE, MALDI-MS, transfection

17

INTRODUCTION

13 14

18 19 20 21 22 23 24 25 26 27

cte

12

rre

11

co

9 10

Alzheimer’s disease (AD) is an irreversible, progressive neurodegenerative disorder causing dementia [1]. The disorder is pathologically characterized by accumulation of amyloid plaques in the brain parenchyma and the main neurotoxic component responsible for the formation of these plaques, which in turn leads to neurodegeneration, is amyloid-␤ (A␤) peptides [2]. A␤ is one of the proteolytic fragments cleaved from the amyloid-␤ protein precursor (A␤PP) by two proteases, ␤- and ␥-secretase [3]. Although extensive studies have

Un

8

dA

15

Abstract. Amyloid-␤ protein precursor intracellular domain (AICD), which exerts intracellular effects by interacting with proteins involved in a plethora of biological processes, is a key player behind the pathophysiology of Alzheimer’s disease (AD). Keeping in mind that overwhelming presence of AICD would mimic AD-like conditions in neuroblastoma cell lines, we hypothesized alteration in the proteomic expression pattern in these cells in the presence of AICD compared to their normal proteome. The rationale behind the study was to distinguish between symptomatic pathophysiological effects as opposed to any artifactual consequence due to protein overload in the cell lines. Using 2D-DIGE analysis and MALDI-MS identifications in neuro2A (mouse) and SHSY5Y (human) cell lines, we have identified several proteins belonging to different functional classes and involved in several biological pathways including protein folding, cytoskeletal dynamics, metabolism, and stress. Many of these were being upregulated or downregulated due to AICD effects and could be correlated directly with AD phenotypes.

7

1 These

authors contributed equally to this manuscript. ∗ Correspondence to: Debashis Mukhopadhyay, Associate Professor, Structural Genomics Division, Saha Institute of Nuclear Physics. Postal address: 1/AF, Bidhannagar, Kolkata-700064, WB, India. Tel.: +91 33 2337 5345; Fax: +91 33 2337 4637; E-mail: [email protected].

been done with A␤ fragment, understanding the complexity of the disease is far behind expectation and search for new players has become a necessity [4]. The A␤PP intracellular domain (AICD), one of the C-terminal fragments of A␤PP, is one such potential candidate that plays an important role in determining cell fate and neurodegeneration [5]. AICD is produced in same amount like A␤ in the amyloidogenic pathway resulting from the same secretase cleavage, and the amino acid sequence of AICD is more conserved through evolution than that of A␤ [6], indicating the possibility of functional involvement of this fragment of A␤PP in the disease pathway. AICD expression is prevalent in AD. Earlier studies exploited AICD-overexpression to mimic AD-like conditions in both cell lines and transgenic mouse models [7, 8], and there have always been concerns about artifactual effects due to overexpression. To resolve

ISSN 1387-2877/13/$27.50 © 2013 – IOS Press and the authors. All rights reserved

28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45

53 54 55 56 57 58 59 60 61

62

63 64

65 66 67 68 69 70 71 72 73 74 75 76 77 78 79 80 81 82 83 84 85 86 87 88 89 90 91 92 93

MATERIALS AND METHODS Neuro2A and SHSY5Y cell culture, transfection of AICD, and protein extraction

roo f

52

or P

51

considered as the AICD-overexpressed proteome of these two cell lines. The corresponding control set of proteome was prepared following the same protein extraction protocol from the control N2A and SHSY5Y cells where transfections were carried out only with empty GFP-C1 vector, i.e., without the expression of AICD. This scramble was used to rule out modifications due to manipulation rather than resulting from AICD overexpression. To show that overexpression of AICD really took place in AICD-GFP transfected cells compared to only GFP transfected cells, western blot with anti-AICD antibody was done (Supplementary Fig. 4) for these two samples and the blots showed that the transfection efficiency and corresponding AICD overexpression was very high in AICD-GFP transfected cells compared to GFP-only transfected cells. Protein quantification was done with the dissolved protein samples by Bradford protein assay using BSA as standard. CyDye labeling

dA

50

The nucleotide sequence of the AICD protein (47 amino acids) was previously cloned in our lab into the mammalian fluorescent expression vector pEGFP-C1 (purchased from Clontech, USA), and this construct (AICD-GFP) was used for transfection into the mammalian neuroblastoma cells (both N2A and SHSY5Y). In the pEGFP-C1 vector, the multiple cloning site (MCS) is followed by GFP coding sequence under the same starting ATG. So genes cloned into the MCS have been expressed as fusion to C terminus of GFP protein. The vector backbone also contains an SV40 origin for replication in mammalian cells and an SV40 early promoter under which the synthesis of mRNA and further expression of the fusion protein occurs. Mouse neuroblastoma Neuro2A (N2A) and human neuroblastoma SHSY5Y cells were grown at 37◦ C in a humidified 5% CO2 incubator in DMEM culture media (Himedia) supplemented with 10% FBS (Biowest). AICD-GFP (4 ␮g in each 90 mm culture plate) was transfected in both the cell lines when it was 75% confluent. 24 h post-transfection, the cell pellets were collected and washed with PBS. The cell pellets were resuspended in lysis buffer (50 mM Tris/Cl, pH 7.5, 1 mM EDTA, 150 mM NaCl, 2 mM PMSF, 0.5% NP-40, and 1X protease inhibitor cocktail, Pierce) and rapid freezing and thawing was carried out to rupture the cell walls. After centrifugation at 13,000 RPM for 15 min at 4◦ C, the supernatant containing the whole cell proteins was collected. This was

cte

49

rre

48

this, we used AICD-overexpressed N2A and SHSY5Y cells to mimic the AD-like condition and identify the differentially expressed proteins due to AICD transfection into those cells. All the proteins we obtained in this study may not directly relate to or influence AD pathogenesis, but others are directly related functionally with the disease. We have shown previously the involvement of AICD in physical interaction with proteins with several different classes of molecular functions using in vitro pull-down experiments with N2A cells [9]. Involvement of these affected proteins in the disease condition is supposed to be mediated by their interactions with AICD, either directly or indirectly. This study is the first approach to understand and compare the effects of AICD on the neuroblastoma proteome from two different organisms.

co

47

Un

46

A. Chakrabarti et al. / AICD-Dependent Altered Neuroblastoma Proteome

uth

2

Both AICD-transfected and control samples were salt depleted using acetone precipitation method. The protein extracts were mixed with 1 : 4 (v/v) acetone, incubated for 3 h at −20◦ C with vortexing in between, and centrifuged at 13,000 rpm for 15 min. Protein precipitates were dissolved in 20 ␮l DIGE labeling buffer (7 M urea, 2 M thiourea, 4% CHAPS, 30 mM Tris pH 8.8, Roche pI cocktail 1 : 100 dilution). In each of the experiments, 50 ␮g of transfected protein and 50 ␮g of control were used. An internal standard was also prepared for each DIGE experiment taking 25 ␮g protein each of the transfected and control proteins, dissolved in 10 ␮l of DIGE buffer, and then pooled. N-hydroxysuccinimide cyanine dyes (CyDyes, Amersham Biosciences, Uppsala, Sweden) were reconstituted in ice, with DMF and added to the protein samples such that there was 200 pM of CyDye per 50 ␮g protein sample with Cy5, Cy3, and Cy2 being used to label the transfected, control and internal standard samples respectively and incubated in ice for 30 min with gentle mixing every 10 min. The reaction was stopped with 10 mM lysine solution after 30 min of labeling. All reactions with the CyDyes were carried out in dark. Post reaction, all labeled samples were mixed up and the total volume was made up to 300 ␮l with rehydration buffer (8 M urea, 4% CHAPS, 2% DTT, 2% ampholyte pH 3–10, and 30 mM Tris/HCl pH 8.5). 17 cm IPG strip (pH 4–7) rehydration and subsequent steps for 2D-gel run were carried out in the dark to avoid quenching of CyDyes.

94 95 96 97 98 99 100 101 102 103 104 105 106 107 108 109 110 111 112

113

114 115 116 117 118 119 120 121 122 123 124 125 126 127 128 129 130 131 132 133 134 135 136 137 138 139 140 141 142 143

A. Chakrabarti et al. / AICD-Dependent Altered Neuroblastoma Proteome

152 153 154 155 156 157 158 159 160 161 162 163 164 165 166 167 168 169

170

171 172 173 174 175 176 177 178 179 180 181 182 183 184 185 186 187 188 189 190 191 192

roo f

151

or P

150

uth

149

Western blot

For validation of differential expression of eight of the proteins, western blot experiments were performed to compare the expression of peroxiredoxin 2, ␣-Tubulin, vimentin, HSPA8, 14-3-3, Pin1, PDI, and stathmin in the proteome of AICD-overexpressed cells and control cells with GFP-only transfection. Cells were transfected separately with AICD-GFP and GFP-only and 24 h post-transfection, proteins were extracted from both the transfected cells and compared. Amount of proteins from each sample run in each western blot experiment was 30 ␮g and for validation of each protein, the experiment was performed at least twice. Antibodies used for this study are as follows: mouse monoclonal anti-peroxiredoxin-2 (ab16748), mouse monoclonal anti-␣-tubulin (ab7291), mouse monoclonal anti-vimentin (V6389), rabbit monoclonal anti-HSPA8 (ab51052), rabbit polyclonal anti-14-33 (ab9063), mouse monoclonal anti-Pin1 (sc46660), rabbit monoclonal anti-PDI (3501S), and rabbit monoclonal anti-stathmin (ab52630). GAPDH was used as loading control and rabbit polyclonal anti-GAPDH antibody (ab9485) was used for this purpose. Densitometry analyses were performed using Versa doc (BioRad) and t-test p-values were calculated using Origin software.

dA

148

ative resolution of 50 (full width at half maximum) and data from 3000–5000 laser shots were collected. Spectral data was analyzed with GPS ExplorerTM version 3.6 (Applied Biosystems). Peptide identification was based on MASCOT database scoring algorithm from Swiss-Prot and NCBI protein databases using search settings of: single missed tryptic cut, fixed carbamidomethylation, variable methionine oxidation and N-terminal acetylation and 150 ppm mass accuracy. Autolytic tryptic peaks were excluded in the Mascot search parameter and p < 0.05 was taken to be significant during identification. Since we had taken a gel-based proteomics approach, we could identify from the protein list given by mascot server, the specific protein in a particular spot using the hint of molecular weight and pI.

cte

147

MALDI-MS identification

The excised spots from the DIGE gels were destained using destaining buffer suitable for different staining procedures and then tryptic digestion was performed overnight with the spots using In-Gel tryptic Digestion Kit (Pierce, USA). The digestion mixture was lyophilized in Heto vacuum centrifuge (Thermo) and resuspended in 50% Acetonitrile (ACN) and 0.1% Trifluoroacetic acid (TFA) solution. The dissolved samples were mixed 1 : 1 (v/v) with ␣-Cyano-4hydroxycinnamic acid (CHCA) matrix and spotted on the MALDI plate and air dried. MALDI analyses were performed with 4700 Proteomic Analyzer MALDI-TOF/TOF (Applied Biosystems) for identification of the protein spots. Peptide mass fingerprinting data was acquired in positive MS reflector mode using fixed laser intensity of 5500 with 2000–3000 laser spots in the range 800–4000 Da, signal to noise ratio 10 and mass exclusion tolerance of 150 ppm. Internal calibration was done in the instrument with minimum signal to noise of 20 and mass tolerance of ±300 ppm taking monoisotopic peaks only. The isolation of peptides of interest was done at a rel-

rre

146

After rehydration, isoelectric focusing (IEF) was performed according to the following protocol: 250 V for 35 min, 1000 V for 1 h, 10000 V for 8 h, and 10000 V up to 65000 Vh. After the first dimension (IEF), IPG strips were equilibrated for 10 min in reducing equilibration buffer (50 mM Tris/HCl pH 8.8, 6 M urea, 30% glycerol, 2% SDS, and 50 mM DTT) followed by 15 min in alkylating equilibration buffer containing 2.5% (w/v) iodoacetamide instead of DTT. Equilibrated IPG strips were run in 12% SDS-PAGE. The gels from each 2D-DIGE experiment was scanned with a Typhoon imager (version 3.0) at excitation/emission values of 488/520 nm for Cy2 dye, 532/580 nm for Cy3 dye, 633/670 nm for Cy5 dye. Gel images were analyzed with DeCyder software (version 6.5, Amersham Biosciences) using the automated spot detection algorithm of Differential In-gel Analysis (DIA) module. A spot was considered as differentially expressed whenever the fold change in DIA was larger than +1.5 or smaller than −1.5 and if t-test p-value were less than 0.05. For each DIGE experiment, post-staining of the gel was done with sypro ruby and the differentially expressed spots as determined by DeCyder software were excised using an automatic spot-cutter (Bio-Rad) for MALDI-MS analysis.

co

145

2D-DIGE analysis

Un

144

3

RESULTS Differentially expressed proteins in N2A and SHSY5Y cells Figure 1 represents differentially expressed N2A proteome upon AICD transfection. Overall, 41

193 194 195 196 197 198 199 200 201 202 203 204 205 206 207 208

209

210 211 212 213 214 215 216 217 218 219 220 221 222 223 224 225 226 227 228 229 230 231 232 233 234

235

236 237

238 239

4

A. Chakrabarti et al. / AICD-Dependent Altered Neuroblastoma Proteome

dA

uth

or P

roo f

sifications of these altered proteins according to PANTHER are represented as a pie chart in the Supplementary Materials. Transfection of SHSY5Y cells with AICD and subsequent 2D-DIGE analyses, as represented in Fig. 2, revealed that several proteins showed altered expression pattern. 25 proteins were upregulated and 20 downregulated among the 45 differentially expressed proteins upon AICD transfection. The spot numbers in this figure is marked according to the spot numbers given in Table 2. Again, PANTHER classification of these altered proteins revealed them to be involved in a variety of molecular functions and biological pathways (Supplementary Materials). It was clear that the altered proteins from both the cell lines represented similar patterns of molecular functional classes involved in similar type of biological pathways.

241 242 243 244 245 246 247 248 249 250 251 252 253 254 255 256

different proteins from N2A cells were found to be differentially expressed due to AICD overexpression. Among them, 21 proteins were upregulated and 20 were downregulated in AICD-GFP transfected cells compared to GFP-transfected control cells. The spot numbers in the figure, as marked match with the spot numbers given in Table 1. PANTHER classification of these proteins revealed that the proteins belonged to several protein classes involved in biological processes having a large spectra of molecular functions. The majority of the proteins possess catalytic activity, binding, and structural roles. The most prevalent biological processes involved cell communications, cell cycle, and cellular component organization. Additionally, some differentially expressed proteins were also involved in immune response as expected in a stressed condition. Clas-

Un

240

co

rre

cte

Fig. 1. Representative 2D-DIGE image showing differential proteomic expression of N2A cells due to AICD-GFP transfection. The whole cell proteome of AICD-GFP transfected cells were labeled with Cy5 and that of the GFP-only transfected cells were labeled with Cy3. The internal standard containing equal amount of both the proteome was labeled with Cy2. In the merged image, after DeCyder analyses, the spots appearing redder are upregulated and the spots that are greener are downregulated in AICD-transfected condition. The yellow spots were with similar expression patterns. The identified proteins showing differential expressions (both upand downregulation) were marked according to the numbers given in Table 1.

Fig. 2. Representative 2D-DIGE image showing differential proteomic expression of SHSY5Y cells due to AICD-GFP transfection. The whole cell proteome of AICD-GFP transfected cells were labeled with Cy5 and that of the GFP-only transfected cells were labeled with Cy3. The internal standard containing equal amount of both the proteome was labeled with Cy2. In the merged image, after DeCyder analyses, the spots appearing redder are upregulated and the spots that are greener are downregulated in AICD-transfected condition. The yellow spots were with similar expression patterns. The identified proteins showing differential expressions (both upand downregulation) were marked according to the numbers given in Table 2.

257 258 259 260 261 262 263 264 265 266 267 268 269 270 271 272 273

A. Chakrabarti et al. / AICD-Dependent Altered Neuroblastoma Proteome

5

Table 1 Differentially expressed N2A proteins upon AICD transfection

21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41

pI

seq cov

Peptides matched

Score

Status

83535.1 72493.5

4.95 5.07

40% 58%

50 54

163 590

up down

P11142 P27797 P07437 P68361 Q9C0J8 O75944 Q99798 Q9NVU0 Q9NX65 P06576

71086.2 48285.8 49370.5 50815.7 146235.3 65944.5 86126 79847.4 54893.7 56524.6

5.37 4.29 4.71 4.94 9.24 7.96 7.62 6.05 9.03 5.26

56% 35% 39% 47% 33% 41% 39% 23% 26% 42%

48 27 36 28 65 49 52 24 17 25

241 167 261 83 58 190 181 53 65 99

down down down down up up up down up down

P49368 P06733 Q6M063 P06733 P60709 P04075 P00558 P40925

60364.3 47487.4 59829.6 47515.4 41709.7 39264.3 44455.1 36272

6.1 7.01 9.16 7.01 5.29 8.39 8.3 6.89

31% 53% 38% 56% 50% 36% 38% 35%

27 44 38 49 20 16 17 19

114 68 127 108 284 63 90 262

up up up down down up up up

Q9NYQ7 P11766 Q04917 P32119 P62937 P18669 P63244 P06748 P63241

uth

or P

roo f

MW

P08238 P11021

dA

13 14 15 16 17 18 19 20

Un

3 4 5 6 7 8 9 10 11 12

Accession No.

363022 41574.3 28172 21843.3 17999.9 28654.8 35088.5 32554.8 16690.4

6.22 7.83 4.76 6.84 7.68 6.75 7.6 4.64 5.08

15% 36% 63% 47% 76% 63% 67% 29% 84%

61 33 41 15 29 33 47 7 17

68 63 80 81 289 101 138 66 226

down down up up up up down down up

P16949 P25398 P15531 P28074

17160.9 14385.4 17137.7 22882.3

5.77 6.36 5.83 8.66

29% 62% 59% 33%

4 11 16 13

61 78 153 61

down down up down

O14904 Q9UF56 P38646

40294 41961.2 73634.8

9.08 7.81 5.87

31% 37% 41%

19 23 34

52 60 69

down down up

P10809 Q13087 P31948 P08670 P35232

61016.4 57080.7 62599.4 53619.1 29785.9

5.7 4.76 6.4 5.06 5.57

48% 51% 56% 51% 50%

41 41 56 26 20

53 72 76 157 313

up up up down down

cte

HSP90 ␤ dnaK type molecular chaperone HSPA5/GRP78 HSPA8 Calreticulin Precursor Tubulin ␤ Tubulin ␣ WD-repeat containing protein Aconitase Aconitase hydratase DNA-directed RNA polymerase III Zinc finger protein 434 ATP synthase ␤ chain, mitochondrial precursor T-complex protein 1, ␥ subunit Phosphopyruvate hydratase H+-transporting two-sector ATPase ␣-Enolase Actin-␤ Fructose bisphosphate aldolase A Phosphoglycerate kinase 1 Malate dehydrogenase, mitochondrial precursor Protocadherin Alcohol dehydrogenase 5 14-3-3η Peroxiredoxin 2 Peptidyl-prolyl cis-trans isomerase Phosphoglycerate mutase Guanine nucleotide binding protein B23 Nucleophosmin Eukaryotic translation initiation factor 5A Stathmin 40 S ribosomal protein S12 Nucleoside diphosphate kinase A Proteasome subunit ␤, type 5 precursor Wnt-9a protein precursor F-box/LRR repeat protein 17 Stress 70 protein/GRP75, mitochondrial precursor HSP60 Protein disulfide isomerase Stress-induced phosphoprotein 1 Vimentin Prohibitin

rre

Protein name

1 2

co

Spot No.

Note: Status is represented as expression in AICD-overexpressed condition compared to that in control. ‘up’ means upregulation of at least 1.5 fold and ‘down’ means at least 1.5 fold downregulation in AICD-transfected condition.

274 275

276 277 278 279 280

Comparison of the altered proteomic expression in N2A and SHSY5Y Comparison of the proteins that were found to be altered in N2A and SHSY5Y due to AICD overexpression revealed some similarity of alteration in both the paradigms. 21 proteins were common in showing differential expression in both the cell lines. These

proteins are listed in Table 3 with short indication of their function along with their alteration pattern (upregulation or downregulation) as a result of AICD transfection. The most prevalent protein class to which the altered proteins from both cell lines belonged to is molecular chaperone. Six proteins out of 21 (∼28.6%), namely, HSP90 ␤, HSPA8, GRP78, HSP60, 14-3-3 protein, and

281 282 283 284 285 286 287 288

6

A. Chakrabarti et al. / AICD-Dependent Altered Neuroblastoma Proteome Table 2 Differentially expressed SHSY5Y proteins upon AICD transfection

25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45

pI

seq cov

Peptides matched

Score

Status

41765.8 53651 61016 72071.2

5.3 5.19 5.7 5.03

46% 51% 57% 51%

26 26 49 46

410 157 261 322

down down up up

Q5CAQ5 Q2VPJ6 P55072 P14314 Q6IAW5 P08865 P06748 P09493 P06753 Q07021

92282 68328.8 89134.7 59258.8 36995.6 32702 32554.8 32688.7 29026.8 31342.6

4.7 5.11 5.14 4.34 4.5 4.79 4.64 4.69 4.73 4.74

51% 50% 52% 27% 28% 61% 40% 49% 71% 29%

56 39 48 23 21 27 24 22 32 10

310 243 138 146 71 148 209 138 237 199

down up down down up up up down down down

P61981 P11142 Q13087 O95672 P02992

28153.9 70854.2 56746.8 87761.5 49510.2

4.8 5.37 5.98 6.56 7.26

49% 56% 48% 12% 41%

24 40 24 10 16

202 288 173 57 109

up down up up up

38448.8 26521.7 249154 38169 53881.2

7.56 6.51 6.49 6.2 5.7

49% 68% 11% 32% 26%

34 44 43 13 12

81 111 51 76 62

down down down down down

P06733 P04179

47139.3 22190.2

7.01 6.86

40% 47%

12 9

71 70

down up

A4D1P6

77165.5

6.28

20%

15

62

up

P32119 Q06830 Q5JC45 CAC88315 P09382 P62937 P36969

21843.3 18963.7 48654.9 19217.5 14592.1 17959.8 22113

6.84 6.41 5.26 7.82 5.33 7.74 8.64

47% 52% 34% 64% 61% 74% 56%

15 14 23 23 16 26 18

70 109 67 284 148 115 43

up up up down up up up

P40926 P18669 P00558 Q15293 P04075 P14625 P08238 P16949 Q9UNM1 P15531 O75944

35508.8 28654.8 44455.1 38866.2 39264.3 92411.3 83081.1 17160.9 10288.6 17137.7 65945

8.92 6.75 8.3 4.86 8.39 4.76 4.97 5.77 8.98 5.83 7.96

46% 27% 38% 33% 22% 51% 39% 35% 47% 55% 40%

20 8 17 17 9 56 42 6 13 17 41

239 62 80 58 64 310 243 70 187 134 180

up up up up up up up down down down down

P07355 P60174 Q96BY6 Q8TE01 P05091

or P

roo f

MW

P63261 P08670 P10809 P11021

rre

20 21 22 23 24

co

15 16 17 18 19

Un

5 6 7 8 9 10 11 12 13 14

Accession No.

uth

Actin ␥ Vimentin HSP60 dnaK type molecular chaperone HSPA5/GRP78 Tumor rejection antigen HSP90AA1 TER ATPase Glucosidase 2, ␤-subunit precursor Calumenin 40 S ribosomal protein SA B23 Nucleophosmin Tropomyosin 1 alpha chain Tropomyosin 3 isoform 2 Complement component 1, Q-binding protein 14-3-3␥ HSPA8 Protein disulfide isomerase Endothelin converting enzyme-like1 Elongation factor Tu, mitochondrial precursor Annexin A2 Triose phosphate isomerase Dedicator of cytokinesis protein-10 DERP12 Mitochondrial aldehyde dehydrogenase ␣-Enolase Superoxide dismutase [Mn], mitochondrial WD repeat containing protein 91/ HSPC049 Peroxiredoxin2 Peroxiredoxin1 Transformation related protein2 Seq. 9 from patent WOO164904 Galactoside protein/Galectin 1 Peptidyl-prolyl cis-trans isomerase Glutathione peroxidase, mitochondrial precursor Mitochondrial Malate dehydrogenase Phosphoglycerate mutase Phosphoglycerate kinase1 Reticulocalbin1 precursor Fructose bisphosphate aldolase A Endoplasmin precursor HSP90 ␤ Stathmin Chaperonin 10-related protein Nucleoside diphosphate kinase A Aconitase

dA

Protein name

1 2 3 4

cte

Spot No.

Note: Status is represented as expression in AICD-overexpressed condition compared to that in control. ‘up’ means upregulation of at least 1.5 fold and ‘down’ means at least 1.5 fold downregulation in AICD-transfected condition.

289 290 291 292

B23 nucleophosmin, represented this class. The proteins vimentin and actin were from the cytoskeletal protein class. Vimentin was also represented in structural protein class. Several proteins were found to have

catalytic activity and they represented different protein classes like isomerases (phosphoglycerate mutase, protein disulfide isomerase, and peptidyl-prolyl cistrans isomerase), kinases (phosphoglycerate kinase

293 294 295 296

A. Chakrabarti et al. / AICD-Dependent Altered Neuroblastoma Proteome

7

Table 3 List of proteins with altered expression in both the paradigms along with their molecular functions Molecular chaperone Molecular chaperone inside ER Molecular chaperone; acts as repressor of transcriptional activation Pre-mRNA 3’ end processing Citric acid cycle enzyme Glycolysis enzyme; takes part in various processes like growth control, hypoxia tolerance and allergic response Structural constituent of cytoskeleton; involved in various types of cell motility Key role in glycolysis and gluconeogenesis; may also function as scaffolding protein Glycolysis enzyme; also acts as primer recognition protein for polymerase Oxidoreductase function in TCA cycle Adaptor for various signalling pathway proteins; have chaperone activity Involved in redox regulation of cells; have oxidoreductase and peroxidase activity Has isomerase activity; also accelerate folding of proteins Intramolecular transferase activity Protein chaperone involved in rRNA metabolism, ribosome biosynthesis etc Involved in regulation of microtubule filament system by destabilizing microtubules- prevents assembly and promotes disassembly Required for assembly/stability of 40S ribosomal subunit; nucleic acid binding Nucleotide kinase; involved in cell proliferation, differentiation and development; also required for neural development, patterning and cell fate determination Molecular chaperone, implicated in mitochondrial protein import and macromolecular assembly Involved in protein modification process with disulfide isomerase activity Structural constituent of cytoskeleton

Actin Fructose bisphosphate aldolase A Phosphoglycerate kinase1 Malate dehydrogenase 14-3-3 Peroxiredoxin2 Peptidyl-prolyl cis-trans isomerase

Stathmin

40S ribosomal protein

HSP60 Protein disulfide isomerase Vimentin

rre

Nucleoside diphosphate kinase A

cte

dA

Phosphoglycerate mutase B23 Nucleophosmin

uth

WD-repeat containing protein Aconitase ␣-Enolase

Status in N2A

Status in SHSY5Y

up down down

up up down

up up down

up down down

roo f

Molecular function

HSP90 ␤ HSPA5/GRP78 HSPA8

or P

Protein name

down

down

up

up

up

up

up up

up up

up

up

up

up

up down

up up

down

down

down

up

up

down

up

up

up

up

down

down

298 299 300 301 302 303 304 305 306 307 308 309 310 311 312

and nucleoside diphosphate kinase), and lyases (fructose bisphosphate aldolase). Phosphoglycerate kinase and nucleoside diphosphate kinase also possessed transferase activity. These proteins have their major roles in different metabolic pathways. The proteins 40S ribosomal protein and WD-repeat containing protein have nucleic acid binding property. Peroxiredoxin 2 and malate dehydrogenase belonged to the class oxidoreductase. Although the pattern of alterations (upregulation or downregulation) for most of the proteins from both cell lines was similar, some of the proteins showed a reverse pattern in their change in expression upon AICD overexpression. For example, the proteins GRP78, WD-repeat containing protein, B23 nucleophosmin, and 40S ribosomal protein showed downregulation in

Un

297

co

Note: “up” and “down” represents upregulations and downregulations in AICD-transfected condition.

N2A but upregulation in SHSY5Y upon AICD transfection. On the other hand, the proteins aconitase and nucleoside diphosphate kinase showed upregulation and downregulation in N2A and SHSY5Y, respectively. 3D expression patterns for 12 proteins found to show similar patterns of alteration in both cell lines upon AICD transfection are represented in Supplementary Fig. 3. Validation of a few differential expression results Eight of the proteins showing altered expression in DIGE experiments were further validated by western blot. All the western blot validations were carried out in SHSY5Y cells. Peroxiredoxin 2 (PRDX2), found to be upregulated by 3.23 fold in

313 314 315 316 317 318 319 320

321

322 323 324 325 326

A. Chakrabarti et al. / AICD-Dependent Altered Neuroblastoma Proteome

uth

or P

roo f

8

330 331 332 333 334 335 336 337 338 339 340 341 342 343 344 345

346

347 348 349

rre

329

western blot which was similar to the observations in DIGE experiments and ␣-tubulin and vimentin, both downregulated in DIGE experiments, were also shown to have downregulations in western blots (by 4.19-fold and 2.17-fold, respectively). Although the protein ␣-tubulin was identified to be differentially expressed only in N2A by DIGE experiments, validation of its downregulation in SHSY5Y represents the limitation of the high-throughput proteomic identification. Other validated proteins found to be upregulated were protein disulfide isomerase (PDI), peptidyl-prolyl cis-trans isomerase (Pin1), and 14-3-3 (upregulated by 3.37-, 1.7-, and 1.68-fold, respectively). Stathmin and HSPA8 were also validated which showed downregulation (1.64- and 1.27-fold, respectively) due to AICD overexpression. Figure 3 represents the western blots for validation of these eight proteins showing altered expression due to AICD transfection.

co

328

Un

327

cte

dA

Fig. 3. Western blots for validation of eight of the proteins altered upon AICD transfection. Peroxiredoxin 2, PDI, Pin1, and 14-3-3 were found to be upregulated, and ␣-tubulin, vimentin, stathmin, and HSPA8 were downregulated, which was similar as obtained in DIGE experiments. All eight proteins showed significant alterations. P-values for PRDX2, PDI, Pin1, 14-3-3, ␣-tubulin, vimentin, stathmin, and HSPA8 were found to be 0.07, 0.05, 0.007, 0.03, 0.01, 0.05, 0.01, and 0.002, respectively, indicated with ‘*’ marks in the bars showing the fold change in protein expression with AICD-GFP transfected condition with respect to that for the GFP-only transfected condition.

DISCUSSIONS Our study is an attempt to decipher the effect of AICD-overexpression in two different neuroblastoma cell lines at the proteome level. The proteins which

show altered expression levels belong to several protein classes participating in different molecular functions. One major class of proteins which showed differential expression upon AICD transfection was cytoskeletal or structural proteins. Among them, actin and vimentin showed altered expression in both cell lines. Disruption of actin dynamics is already reported due to induction of AICD expression in neuronal cells [10]. Some isoforms of tubulin were found to be differentially expressed in N2A cells, and tropomyosin ␣ showed altered expression in SHSY5Y cells. We also obtained 40S ribosomal subunits, though different subunits in these two cell lines, showing altered expression in our study. Loss of structural integrity is a wellestablished event in neurodegenerative diseases like AD [11]. Altered expression of the structural proteins due to AICD overload, therefore, is definitely an important observation. We previously showed vimentin as one of the several interactors of AICD [9]. Alteration of vimentin found in our study further strengthens the role of AICD in cytoskeletal restructuring. Stathmin, downregulated in both cell lines in our study, functions as a regulatory protein of microtubule dynamics and was found to be associated with neurofibrillary tangles in brains of AD patients [12]. This protein is also reported to be downregulated by AICD-Fe65-Tip60 complex in

350 351 352 353 354 355 356 357 358 359 360 361 362 363 364 365 366 367 368 369 370 371 372 373 374 375

A. Chakrabarti et al. / AICD-Dependent Altered Neuroblastoma Proteome

384 385 386 387 388 389 390 391 392 393 394 395 396 397 398 399 400 401 402 403 404 405 406 407 408 409 410 411 412 413 414 415 416 417 418 419 420 421 422 423 424 425 426 427

roo f

383

or P

382

uth

381

dA

380

level was also found to be decreased in adult brain with Down syndrome and AD [26]. It is generally perceived that there might be a link between Down syndrome and overproduction of AICD that leads to an AD-like condition [27]. Once again, decreased expression of stathmin due to AICD overexpression might have some role to link these two diseases. Other proteins which were reported to be altered during aging and also showed up- or downregulation in our study include triosephosphate isomerase, superoxide dismutase, annexin, enolase, and protein disulfide isomerase, among others [28]. Several heat shock proteins (HSPs), e.g., HSP90␤, GRP78, GRP75, HSPA8, HSP60, and T-complex protein1, were found to show altered level of expression in our experiments conforming with a previous study with AD brain [29]. AD-like neurodegenerative features are the result of gain of toxic functions of misfolded proteins/peptides (A␤ and tau), and aberrant chaperone activities are thought to be responsible for the shift of balance between folded to misfolded proteins [30]. Several studies have been conducted to explain the role of chaperones, mainly the HSPs, in AD [31, 32]. Our results, therefore, are in accordance with the hypotheses regarding the involvement of chaperones as well as AICD in AD pathophysiology. Although there were some inconsistencies in the direction of change of the HSPs in our study and studies by others, this may be explained by differential response of individual HSPs to a stressed condition [33]. Quite a few metabolic proteins involved in a variety of biochemical pathways including glycolysis, Krebs cycle, oxidative phosphorylation, mitochondrial protein synthesis, etc., showed altered expression in this study hinting that active metabolic changes had occurred due to AICD transfection. In a protein misfolding and aggregation disorder like AD, it is expected that metabolic protein expression should get altered due to changes in the energy need [34]. There is strong evidence in favor of mitochondrial dysfunction contributing to AD phenotype [35–37]. A number of mitochondrial proteins in this study were found to have differential expression in the AICDtransfected condition. An imbalance in mitochondrial bioenergetics was also previously reported due to the presence of AICD [10]. Among these, mitochondrial malate dehydrogenase, superoxide dismutase [Mn], fructose bisphosphate aldolase A, etc., were found to be upregulated in an AICD-overexpressed condition. These proteins were also reported to be upregulated in an earlier study of mitochondrial proteome with mouse model of AD [38]. However, the proteins fructose

cte

379

rre

378

HEK293T cells [13]. Our result suggests the possibility of AICD-mediated role of stathmin in microtubule disorganization during AD progression. Oxidative stress is another feature for the pathogenesis of AD [14–16]. In the present study, we found several antioxidant proteins to be upregulated due to AICD-overexpression. The proteins peroxiredoxin 1, peroxiredoxin 2, and mitochondrial superoxide dismutase, which were observed to be altered (increased) in our study, were also previously reported to show increased expression in different regions of the brain of AD patients [17, 18]. Peroxiredoxin 1 and peroxiredoxin 2 play important antioxidant roles for protecting neurons from peroxide-induced damage. Elevated expression of these proteins (both PRDX1 and PRDX2 in SHSY5Y and PRDX2 in N2A) indicates the involvement of reactive oxygen species in AD pathogenesis [18]. Two other antioxidant proteins, glutathione peroxidase in SHSY5Y and alcohol dehydrogenase in N2A were also showed to have increased and decreased expressions, respectively. Although we did not find any protein involved in autophagy or vesicle formation to show altered expression, several signaling proteins were found to be altered in this study. The protein Pin1 was found to be upregulated in both the cell lines. Of late it has been reported that Pin1 is involved in several signaling pathways as well as in regulation of cell cycle and cell division [19]. Due to its involvement in aging process and age-related disorders, it plays a pivotal role in cancer as well as in AD and can act as an important link between the two [19, 20]. Wnt signaling is increased during normal aging [21] and this pathway is also involved with Pin1 and p53 linking aging, cancer, and AD [22]. We found Wnt-9a protein precursor, involved in developmental processes, having shown decreased expression in N2A cells upon AICD overexpression. This result, along with increased expression of Pin1, strengthen the hypothesis that normal aging and AD are coupled processes and the developmental defects linked with them, as well as cancer, do have some level of commonality. Additionally, AICD-mediated transcriptional activity is promoted by Wnt/␤-catenin signaling [23], further implying the influence of AICD on these pathways. Stathmin was earlier reported to have altered expressions in the neonatal mice brain as well as in old postmortem human brain in comparison to normal adults [24, 25]. This protein was found to be downregulated in this study with overexpression of AICD, and hence, its involvement in the age-dependent decline in neurogenesis as well as in the disease process can be thought to be mediated by AICD. Stathmin

co

377

Un

376

9

428 429 430 431 432 433 434 435 436 437 438 439 440 441 442 443 444 445 446 447 448 449 450 451 452 453 454 455 456 457 458 459 460 461 462 463 464 465 466 467 468 469 470 471 472 473 474 475 476 477 478 479

487 488 489 490 491 492 493 494 495 496 497 498 499 500 501 502 503 504

505

506 507 508 509 510 511

512

513 514 515

516

517

ACKNOWLEDGMENTS

The work was partially funded by the IBOP project, Department of Atomic Energy, Government of India. AC acknowledges Council of Scientific and Industrial Research, Government of India for his fellowship. Authors’ disclosures available online (http://www.jalz.com/disclosures/view.php?id=1909). SUPPLEMENTARY MATERIAL

526 527

[11]

[12]

[13]

[14]

[15]

[16]

[18]

[1] [2] [3]

523

525

[10]

REFERENCES

522

524

[9]

[17]

520 521

[8]

Supplementary material and figures are available in the electronic version of this article: http://dx.doi.org/10.3233/JAD-130695.

518 519

[7]

[4]

Selkoe DJ (2001) Alzheimer’s disease: Genes, proteins, and therapy. Physiol Rev 81, 741-766. Yankner BA (1996) Mechanisms of neuronal degeneration in Alzheimer’s disease. Neuron 16, 921-932. Wilquet V, De Strooper B (2004) Amyloid-beta precursor protein processing in neurodegeneration. Curr Opin Neurobiol 14, 582-588. Muller T, Meyer HE, Egensperger R, Marcus K (2008) The amyloid precursor protein intracellular domain (AICD) as modulator of gene expression, apoptosis, and cytoskeletal dynamics – Relevance for Alzheimer’s disease. Prog Neu-

roo f

486

[6]

robiol 85, 393-406. Raychaudhuri M, Mukhopadhyay D (2007) AICD and its adaptors – In search of new players. J Alzheimers Dis 11, 343-358. Nakayama K, Ohkawaraa T, Hiratochi M, Koh CS, Nagase H (2008) The intracellular domain of amyloid precursor protein induces neuron-specific apoptosis. Neurosci Lett 444, 127131. Ghosal K, Vogt DL, Liang M, Shen Y, Lamb BT, Pimplikar SW (2009) Alzheimer’s disease-like pathological features in transgenic mice expressing the APP intracellular domain. Proc Natl Acad Sci USA 106, 18367-18372. Raychaudhuri M, Mukhopadhyay D (2011) AICD overexpression in Neuro 2A cells regulates expression of PTCH1 and TRPC5. Int J Alzheimers Dis 2011, pii: 239453. Chakrabarti A, Mukhopadhyay D (2012) Novel adaptors of amyloid precursor protein intracellular domain and their functional implications. Genomics Proteomics Bioinformatics 10, 208-216. Ward MW, Concannon CG, Whyte J, Walsh CM, Corley B, Prehn JH (2010) The amyloid precursor protein intracellular domain (AICD) disrupts actin dynamics and mitochondrial bioenergetics. J Neurochem 113, 275-284. Fellgiebel A, Schermuly I, Gerhard A, Keller I, Albrecht J, Weibrich C, Muller MJ, Stoeter P (2008) Functional relevant loss of long association fibre tracts integrity in early Alzheimer’s disease. Neuropsychologia 46, 1698-1706. Jin LW, Masliah E, Iimoto D, Deteresa R, Mallory M, Sundsmo M, Mori N, Sobel A, Saitoh T (1996) Neurofibrillary tangle-associated alteration of stathmin in Alzheimer’s disease. Neurobiol Aging 17, 331-341. Muller T, Schrotter A, Loosse C, Pfeiffer K, Theiss C, Kauth M, Meyer HE, Marcus K (2013) A ternary complex consisting of AICD, FE65, and TIP60 down-regulates Stathmin1. Biochim Biophys Acta 1834, 387-394. Butterfield DA, Lauderback CM (2002) Lipid peroxidation and protein oxidation in Alzheimer’s disease brain: Potential causes and consequences involving amyloid beta-peptideassociated free radical oxidative stress. Free Radic Biol Med 32, 1050-1060. Butterfield DA, Castegna A, Lauderback CM, Drake J (2002) Evidence that amyloid beta-peptide-induced lipid peroxidation and its sequelae in Alzheimer’s disease brain contribute to neuronal death. Neurobiol Aging 23, 655-664. Butterfield DA, Drake J, Pocernich C, Castegna A (2001) Evidence of oxidative damage in Alzheimer’s disease brain: Central role for amyloid beta-peptide. Trends Mol Med 7, 548-554. Kim SH, Fountoulakis M, Cairns N, Lubec G (2001) Protein levels of human peroxiredoxin subtypes in brains of patients with Alzheimer’s disease and Down syndrome. J Neural Transm Suppl 223-235. Krapfenbauer K, Engidawork E, Cairns N, Fountoulakis M, Lubec G (2003) Aberrant expression of peroxiredoxin subtypes in neurodegenerative disorders. Brain Res 967, 152-160. Driver JA, Lu KP (2010) Pin1: A new genetic link between Alzheimer’s disease, cancer and aging. Curr Aging Sci 3, 158165. Lee TH, Pastorino L, Lu KP (2011) Peptidyl-prolyl cis-trans isomerase Pin1 in ageing, cancer and Alzheimer disease. Expert Rev Mol Med 13, e21. Brack AS, Conboy MJ, Roy S, Lee M, Kuo CJ, Keller C, Rando TA (2007) Increased Wnt signaling during aging alters muscle stem cell fate and increases fibrosis. Science 317, 807-810.

or P

485

[5]

dA

484

cte

483

rre

482

bisphosphate aldolase A and superoxide dismutase [Mn] were also reported to be altered in human AD brain in several studies by different groups [39–43]. A recent review from the systems biology aspects of AD discussed the diverse molecular changes that might result in the memory impairment [44] and according to their compilation, a majority of the altered proteins from animal models of AD were involved in cytoskeleton organization, energy metabolism, response to oxidative stress, and chaperone activity, which is highly congruent to our data. Another differential proteomics study with A␤1-42 treatment in differentiated SHSY5Y cells (mimicking the AD condition) revealed similar kinds of molecular functional classes the altered proteins were involved in [45]. It is obvious that several pathways are engaged in AD pathogenesis and the ensemble of proteins obtained in our study as well as in studies by other groups strongly suggests that there must be a complex protein interaction network leading to AD with the short intracellular fragment of A␤PP, the AICD, situated in a central node of this complex network. It is likely that a complex pathway interaction network, rather than a protein interaction network, can explain the molecular mechanism behind AD.

co

481

Un

480

A. Chakrabarti et al. / AICD-Dependent Altered Neuroblastoma Proteome

uth

10

[19]

[20]

[21]

528 529 530 531 532 533 534 535 536 537 538 539 540 541 542 543 544 545 546 547 548 549 550 551 552 553 554 555 556 557 558 559 560 561 562 563 564 565 566 567 568 569 570 571 572 573 574 575 576 577 578 579 580 581 582 583 584 585 586 587 588 589 590 591 592

A. Chakrabarti et al. / AICD-Dependent Altered Neuroblastoma Proteome

600

[24]

601 602 603

[25]

604 605 606 607

[26]

608 609 610 611

[27]

612 613 614

[28]

615 616

[29]

617 618 619 620

[30]

621 622 623

[31]

624 625 626 627

[32]

628 629 630 631

[33]

632 633 634 635 636 637

[34]

[37]

[38]

[39]

[40]

[41]

roo f

599

or P

598

[36]

uth

597

Reddy PH, Beal MF (2008) Amyloid beta, mitochondrial dysfunction and synaptic damage: Implications for cognitive decline in aging and Alzheimer’s disease. Trends Mol Med 14, 45-53. Crouch PJ, Harding SM, White AR, Camakaris J, Bush AI, Masters CL (2008) Mechanisms of A beta mediated neurodegeneration in Alzheimer’s disease. Int J Biochem Cell Biol 40, 181-198. Moreira PI, Duarte AI, Santos MS, Rego AC, Oliveira CR (2009) An integrative view of the role of oxidative stress, mitochondria and insulin in Alzheimer’s disease. J Alzheimers Dis 16, 741-761. Chou JL, Shenoy DV, Thomas N, Choudhary PK, Laferla FM, Goodman SR, Breen GA (2011) Early dysregulation of the mitochondrial proteome in a mouse model of Alzheimer’s disease. J Proteomics 74, 466-479. Sultana R, Boyd-Kimball D, Cai J, Pierce WM, Klein JB, Merchant M, Butterfield DA (2007) Proteomics analysis of the Alzheimer’s disease hippocampal proteome. J Alzheimers Dis 11, 153-164. Muller T, Jung K, Ullrich A, Schrotter A, Meyer HE, Stephan C, Egensperger R, Marcus K (2008) Disease state, age, sex, and post-mortem time-dependent expression of proteins in AD vs. control frontal cortex brain samples. Curr Alzheimer Res 5, 562-571. Omar RA, Chyan YJ, Andorn AC, Poeggeler B, Robakis NK, Pappolla MA (1999) Increased expression but reduced activity of antioxidant enzymes in Alzheimer’s disease. J Alzheimers Dis 1, 139-145. Marcus DL, Strafaci JA, Freedman ML (2006) Differential neuronal expression of manganese superoxide dismutase in Alzheimer’s disease. Med Sci Monitor 12, BR8-B14. Perluigi M, Sultana R, Cenini G, Di Domenico F, Memo M, Pierce WM, Coccia R, Butterfield DA (2009) Redox proteomics identification of 4-hydroxynonenal-modified brain proteins in Alzheimer’s disease: Role of lipid peroxidation in Alzheimer’s disease pathogenesis. Proteom Clin Appl 3, 682-693. Juhasz G, Foldi I, Penke B (2011) Systems biology of Alzheimer’s disease: How diverse molecular changes result in memory impairment in AD. Neurochem Int 58, 739-750. Foldi I, Datki ZL, Szabo Z, Bozso Z, Penke B, Janaky T (2011) Proteomic study of the toxic effect of oligomeric Abeta142 in situ prepared from ‘iso-Abeta1-42’. J Neurochem 117, 691-702.

dA

[23]

[35]

[42]

[43]

cte

596

rre

595

Behrens MI, Lendon C, Roe CM (2009) A common biological mechanism in cancer and Alzheimer’s disease? Curr Alzheimer Res 6, 196-204. Zhou F, Gong K, van Laar T, Gong Y, Zhang L (2011) Wnt/beta-catenin signal pathway stabilizes APP intracellular domain (AICD) and promotes its transcriptional activity. Biochem Biophys Res Comm 412, 68-73. Carrette O, Burkhard PR, Hochstrasser DF, Sanchez JC (2006) Age-related proteome analysis of the mouse brain: A 2-DE study. Proteomics 6, 4940-4949. Chen W, Ji J, Xu X, He S, Ru B (2003) Proteomic comparison between human young and old brains by two-dimensional gel electrophoresis and identification of proteins. Int J Dev Neurosci 21, 209-216. Cheon MS, Fountoulakis M, Cairns NJ, Dierssen M, Herkner K, Lubec G (2001) Decreased protein levels of stathmin in adult brains with Down syndrome and Alzheimer’s disease. J Neural Transm Suppl, 281-288. Ghosh S, Chakrabarti A, Mukhopadhyay D (2012) ”Patch”ing up the neurons: Revival or enervation? Vitam Horm 88, 439-459. Chakrabarti A, Mukhopadhyay D (2012) BRAIN SENESCENCE-OMICS. J Proteins Proteomics 3, 15-29. Yoo BC, Kim SH, Cairns N, Fountoulakis M, Lubec G (2001) Deranged expression of molecular chaperones in brains of patients with Alzheimer’s disease. Biochem Biophys Res Comm 280, 249-258. Barral JM, Broadley SA, Schaffar G, Hartl FU (2004) Roles of molecular chaperones in protein misfolding diseases. Semin Cell Dev Biol 15, 17-29. Koren J, 3rd, Jinwal UK, Lee DC, Jones JR, Shults CL, Johnson AG, Anderson LJ, Dickey CA (2009) Chaperone signalling complexes in Alzheimer’s disease. J Cell Mol Med 13, 619-630. Dou F, Netzer WJ, Tanemura K, Li F, Hartl FU, Takashima A, Gouras GK, Greengard P, Xu H (2003) Chaperones increase association of tau protein with microtubules. Proc Natl Acad Sci USA 100, 721-726. Papassotiropoulos A, Fountoulakis M, Dunckley T, Stephan DA, Reiman EM (2006) Genetics, transcriptomics, and proteomics of Alzheimer’s disease. J Clin Psychiat 67, 652-670. Gomes RA, Franco C, Da Costa G, Planchon S, Renaut J, Ribeiro RM, Pinto F, Silva MS, Coelho AV, Freire AP, Cordeiro C (2012) The proteome response to amyloid protein expression in vivo. PloS One 7, e50123.

co

[22]

594

Un

593

[44]

[45]

11

638 639 640 641 642 643 644 645 646 647 648 649 650 651 652 653 654 655 656 657 658 659 660 661 662 663 664 665 666 667 668 669 670 671 672 673 674 675 676 677 678 679 680 681 682 683

Lihat lebih banyak...

Comentarios

Copyright © 2017 DATOSPDF Inc.