7
A new brain-derived neurotrophic factor transcript and decrease in brain-derived neurotrophic factor transcripts 1, 2 and 3 in Alzheimer’s disease parietal cortex Diego Garzon, Guanhua Yu and Margaret Fahnestock Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, Ontario, Canada Abstract Brain-derived neurotrophic factor (BDNF) supports hippo- campal, cortical and basal forebrain cholinergic neurons, which lose function in Alzheimer’s disease. In Alzheimer’s tissues such as hippocampus and parietal cortex, brain- derived neurotrophic factor mRNA is decreased three- to four- fold compared with controls. However, the molecular mechanism of the down-regulation of BDNF in Alzheimer’s disease is unknown. The human brain-derived neurotrophic factor gene has multiple promoters governing six non-coding upstream exons that are spliced to one downstream coding exon, leading to six different transcripts. Here we report an alternate human splice variant within exon 4I for a total of seven transcripts. Previous brain-derived neurotrophic factor mRNA measurements in Alzheimer’s disease tissue were done using the downstream coding exon present in all tran- scripts. Using RT-PCR primers specific for each upstream exon, we observe a significant decrease in three human brain- derived neurotrophic factor mRNA transcripts in Alzheimer’s disease samples compared with controls. Transcripts 1 and 3 each exhibit a two-fold decrease, and transcript 2 shows a five-fold decrease. There are no significant differences between control and Alzheimer’s disease samples for the other transcripts, including the new splice variant. In rat, both transcripts 1 and 3 are regulated through the transcription factor cAMP response element binding protein, whose phos- phorylation is decreased in the Alzheimer’s disease brain. This could lead to specific down-regulation of the brain- derived neurotrophic factor transcripts shown here. Keywords: brain-derived neurotrophic factor, gene expres- sion, human, mRNA, RT-PCR. J. Neurochem. (2002) 82, 1058–1064. Brain-derived neurotrophic factor (BDNF) is highly expressed and is distributed widely throughout the CNS, specifically in the hippocampal formation, cerebral cortex, and amygdaloid complex (Ernfors et al. 1990; Hofer et al. 1990; Phillips et al. 1990; Wetmore et al. 1990). BDNF promotes the survival and function of hippocampal and cortical neurons (Ghosh et al. 1994; Lindholm et al. 1996; Lowenstein and Arsenault 1996), cholinergic neurons (Alderson et al. 1990; Knusel et al. 1991) and nigral dopaminergic neurons (Hyman et al. 1991; Knusel et al. 1991). BDNF is also important for synaptic transmission and the excitatory properties of these neurons (Patterson et al. 1992; Castren et al. 1993; Dragunow et al. 1993; Kang and Schuman 1995; Scharfman 1997; Osehobo et al. 1999; McLean et al. 2000). Basal forebrain cholinergic, cortical, and hippocampal neurons lose function and synaptic connectivity in Alzhei- mer’s disease (AD) (Coyle et al. 1983; Whitehouse et al. 1982; Cuello and Sofroniew 1984; Etienne et al. 1986; Hefti and Weiner 1986; Mann 1991). This may occur because of a deficit in BDNF in the AD brain. A 3–4-fold reduction in BDNF mRNA has been amply documented in the hippo- campus and parietal cortex (Phillips et al. 1991; Holsinger et al. 2000). Protein levels of BDNF have been shown to decrease in Alzheimer’s disease entorhinal cortex, hippo- campus and temporal, frontal and parietal cortex (Narisawa- Saito et al. 1996; Connor et al. 1997; Ferrer et al. 1999; Hock et al. 2000). However, the transcriptional regulation of the human BDNF gene has not been studied, and so the Received February 20, 2002; revised manuscript received May 10, 2002; accepted May 13, 2002. Address correspondence and reprint requests to Dr Margaret Fahne- stock, Department of Psychiatry and Behavioural Neurosciences, McMaster University, 1200 Main Street West, Hamilton, Ontario, Canada L8N 3Z5. E-mail: [email protected] Abbreviations used: AD, Alzheimer’s disease; BDNF, brain-derived neurotrophic factor; CRE, cAMP response element; CREB, cAMP response element binding protein. Journal of Neurochemistry , 2002, 82, 1058–1064 1058 ȑ 2002 International Society for Neurochemistry, Journal of Neurochemistry , 82, 1058–1064

A new brain-derived neurotrophic factor transcript and decrease inbrain-derived neurotrophic factor transcripts 1, 2 and 3 in Alzheimer's disease parietal cortex

Embed Size (px)

Citation preview

Page 1: A new brain-derived neurotrophic factor transcript and decrease inbrain-derived neurotrophic factor transcripts 1, 2 and 3 in Alzheimer's disease parietal cortex

A new brain-derived neurotrophic factor transcript and decrease

in brain-derived neurotrophic factor transcripts 1, 2 and 3 in

Alzheimer’s disease parietal cortex

Diego Garzon, Guanhua Yu and Margaret Fahnestock

Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, Ontario, Canada

Abstract

Brain-derived neurotrophic factor (BDNF) supports hippo-

campal, cortical and basal forebrain cholinergic neurons,

which lose function in Alzheimer’s disease. In Alzheimer’s

tissues such as hippocampus and parietal cortex, brain-

derived neurotrophic factor mRNA is decreased three- to four-

fold compared with controls. However, the molecular

mechanism of the down-regulation of BDNF in Alzheimer’s

disease is unknown. The human brain-derived neurotrophic

factor gene has multiple promoters governing six non-coding

upstream exons that are spliced to one downstream coding

exon, leading to six different transcripts. Here we report an

alternate human splice variant within exon 4I for a total of

seven transcripts. Previous brain-derived neurotrophic factor

mRNA measurements in Alzheimer’s disease tissue were

done using the downstream coding exon present in all tran-

scripts. Using RT-PCR primers specific for each upstream

exon, we observe a significant decrease in three human brain-

derived neurotrophic factor mRNA transcripts in Alzheimer’s

disease samples compared with controls. Transcripts 1 and 3

each exhibit a two-fold decrease, and transcript 2 shows a

five-fold decrease. There are no significant differences

between control and Alzheimer’s disease samples for the

other transcripts, including the new splice variant. In rat, both

transcripts 1 and 3 are regulated through the transcription

factor cAMP response element binding protein, whose phos-

phorylation is decreased in the Alzheimer’s disease brain.

This could lead to specific down-regulation of the brain-

derived neurotrophic factor transcripts shown here.

Keywords: brain-derived neurotrophic factor, gene expres-

sion, human, mRNA, RT-PCR.

J. Neurochem. (2002) 82, 1058–1064.

Brain-derived neurotrophic factor (BDNF) is highly

expressed and is distributed widely throughout the CNS,

specifically in the hippocampal formation, cerebral cortex,

and amygdaloid complex (Ernfors et al. 1990; Hofer et al.

1990; Phillips et al. 1990; Wetmore et al. 1990). BDNF

promotes the survival and function of hippocampal and

cortical neurons (Ghosh et al. 1994; Lindholm et al. 1996;

Lowenstein and Arsenault 1996), cholinergic neurons

(Alderson et al. 1990; Knusel et al. 1991) and nigral

dopaminergic neurons (Hyman et al. 1991; Knusel et al.

1991). BDNF is also important for synaptic transmission and

the excitatory properties of these neurons (Patterson et al.

1992; Castren et al. 1993; Dragunow et al. 1993; Kang and

Schuman 1995; Scharfman 1997; Osehobo et al. 1999;

McLean et al. 2000).

Basal forebrain cholinergic, cortical, and hippocampal

neurons lose function and synaptic connectivity in Alzhei-

mer’s disease (AD) (Coyle et al. 1983; Whitehouse et al.

1982; Cuello and Sofroniew 1984; Etienne et al. 1986; Hefti

and Weiner 1986; Mann 1991). This may occur because of a

deficit in BDNF in the AD brain. A 3–4-fold reduction in

BDNF mRNA has been amply documented in the hippo-

campus and parietal cortex (Phillips et al. 1991; Holsinger

et al. 2000). Protein levels of BDNF have been shown to

decrease in Alzheimer’s disease entorhinal cortex, hippo-

campus and temporal, frontal and parietal cortex (Narisawa-

Saito et al. 1996; Connor et al. 1997; Ferrer et al. 1999;

Hock et al. 2000). However, the transcriptional regulation of

the human BDNF gene has not been studied, and so the

Received February 20, 2002; revised manuscript received May 10, 2002;

accepted May 13, 2002.

Address correspondence and reprint requests to Dr Margaret Fahne-

stock, Department of Psychiatry and Behavioural Neurosciences,

McMaster University, 1200 Main Street West, Hamilton, Ontario,

Canada L8N 3Z5. E-mail: [email protected]

Abbreviations used: AD, Alzheimer’s disease; BDNF, brain-derived

neurotrophic factor; CRE, cAMP response element; CREB, cAMP

response element binding protein.

Journal of Neurochemistry, 2002, 82, 1058–1064

1058 � 2002 International Society for Neurochemistry, Journal of Neurochemistry, 82, 1058–1064

Page 2: A new brain-derived neurotrophic factor transcript and decrease inbrain-derived neurotrophic factor transcripts 1, 2 and 3 in Alzheimer's disease parietal cortex

mechanism of the decrease in BDNF levels in the AD brain

is not well understood.

In the rat, the BDNF gene has been shown to have four 5¢exons and one 3¢ exon (Timmusk et al. 1993). The four 5¢exons each have upstream promoters and each is individually

spliced to the 3¢ exon (encoding the mature protein) to givefour different transcripts. Downstream of the 3¢ exon thereare two polyadenylation sites, which give two different

length transcripts for each upstream exon, totaling eight

splice variants (Timmusk et al. 1993). The presence of

multiple promoters in the BDNF gene allow for differential

mechanisms of activation and tissue-specific expression in

the CNS (Falkenberg et al. 1992; Metsis et al. 1993; Kokaia

et al. 1994; Timmusk et al. 1995).

The human BDNF gene is structurally similar to the rat

gene (Maisonpierre et al. 1991). The human BDNF gene

contains two additional non-coding exons compared with rat

(4I and 5U), but only one polyadenylation site is present

downstream from the 3¢ coding exon (Aoyama et al. 2001).

The six upstream exons in the human BDNF gene give rise to

six transcripts, although the additional exons produce

transcripts by differential splicing, not additional promoters

(Aoyama et al. 2001). Previous measurements documenting

decreased BDNF mRNA in the AD brain targeted the coding

exon (exon5) present in all transcripts and therefore exam-

ined total BDNF mRNA levels. In this study, we used

RT-PCR with upstream primers specific for each exon to

determine which of the transcripts is responsible for the

reduced BDNF mRNA in AD brain.

Materials and methods

Human post-mortem brain tissue

Parietal cortex tissue samples from normal, neurologically

unimpaired subjects (n ¼ 12; six females, six males) and from

subjects with AD (n ¼ 12; six females and six males) were provided

by the Institute for Brain Aging and Dementia Tissue Repository at

the University of California, Irvine. A diagnosis of AD was

confirmed by pathological and clinical criteria (McKhann et al.

1984; Khatchaturian 1985). Control and AD samples were matched

for age and gender. Tissue was frozen at autopsy and stored at

)80�C until use.

RNA isolation

Total cellular RNA was purified from parietal cortex samples using

TRIzolTM Reagent (Gibco BRL, Burlington, Ontario, Canada)

following the manufacturer’s protocol. Samples exhibiting an

absorbance ratio (260/280) greater than or equal to 1.7 and exhib-

iting strong 28S and 18S ribosomal RNA bands on 0.01-g/mL

agarose gels were used for further analysis.

Primers for BDNF transcripts and b-actin

Human BDNF primer sequences were kindly provided by

Dr Mineyoshi Aoyama, Department of Bioregulation Research,

Nagoya City University Medical School, Mizuho-ku, Nagoya,

Japan. b-Actin primer sequences were previously described (StAmand et al. 1996). Primers were synthesized at the Central Facility

of the Institute for Molecular Biology and Biotechnology (MOBIX)

at McMaster University.

RT-PCR

For determination of transcripts 2 and 5U, RNA samples were

treated with DNaseI (1.0 lL/10 lg total RNA; Ambion, Austin, TX,USA) at 37�C for 30 min, followed by 2 lL of DNase Inactivationreagent. For all other transcripts, RNA samples did not undergo

DNase treatment. Ten micrograms total RNA from human parietal

cortex was reverse transcribed into cDNA using the GeneAmp�RNA PCR kit (Perkin Elmer, Norwalk, CT, USA). PCR was

performed in the GeneAmp PCR system 2400 using 5 lL aliquotsof the reverse transcriptase reaction mixture with 0.35 lM each ofthe 3¢ and 5¢ primers, 16.8 lCi of 33P-dCTP, and 2.5 U AmpliTaqGold (Perkin Elmer). Optimization was performed for all primer sets

to determine an optimal cycle number within the logarithmic phase

of amplification. Cycle optimization for transcripts are as follows,

transcript 1, 35 cycles, transcript 2, 34 cycles, transcript 3, 35 cycles,

transcript 4, 32 cycles, transcript 4I, 38 cycles, transcript 5U, 35

cycles, and b-actin, 22 cycles. The amplification profile included aninitial activation of the Taq polymerase for 12 min at 95�C,denaturation for 30 s at 94�C followed by annealing at 58�C for

30 s, extension at 72�C for 45 s, and a final extension at 72�C for7 min. For b-actin, annealing was at 64�C for 30 s and extension at72�C for 1 min. Four to five independent RT-PCRs were performedfor each primer pair.

Isolation and sequencing of RT-PCR products

Amplified RT-PCR product from transcript 4I gave two bands upon

electrophoresis in a 0.018-g/mL agarose gel in the presence of

ethidium bromide. Single bands were cut from the gel, and DNA

was isolated using QIAquickTM Gel Extraction Kit (Qiagen,

Mississauga, ON, Canada). The isolated bands were sequenced in

both directions, using transcript 4I PCR primers, by the Central

Facility of the Institute for Molecular Biology and Biotechnology

(MOBIX) at McMaster University.

Quantitative and statistical analysis

Ten microliters of each RT-PCR reaction mixture was subjected to

electrophoresis in a 0.018-g/mL agarose gel and analyzed by

phosphorimagery using ImageQuant software (Molecular Dynam-

ics, Sunnyvale, CA, USA). Quantitation was accomplished by

placing rectangular cursors of fixed dimensions over each band and

measuring pixel density for each sample, with local background

subtraction. Four to five separate RT-PCR experiments were

performed for each primer pair on each subject; the mean value of

these experiments was used in the statistical analysis (Table 1). For

transcripts with extremely low expression levels as indicated by

pixel values, specifically transcripts 2, 4I, and 5U, only two RT-PCR

experiments were averaged for each subject. For statistical analysis

of group differences in transcript expression, a two-way ANOVA was

used [group (control versus AD subjects) · transcripts (1, 2, 3, 4, 4I,4Ia, and 5U)], followed by two-tailed t-tests to determine signifi-

cance between groups for each transcript. Box plot analysis

identified extreme outliers in transcripts 2, 3 and 5U (Table 1).

These outliers were eliminated from the two-way ANOVA, post-hoc

Brain-derived neurotrophic factor transcripts 1059

� 2002 International Society for Neurochemistry, Journal of Neurochemistry, 82, 1058–1064

Page 3: A new brain-derived neurotrophic factor transcript and decrease inbrain-derived neurotrophic factor transcripts 1, 2 and 3 in Alzheimer's disease parietal cortex

Tab

le1

Results

of

RT

-PC

Rexperim

ents

perf

orm

ed

for

each

prim

er

pair

on

each

subje

ct

Tra

nscript

1S

EM

Tra

nscript

2S

EM

Tra

nscript

3S

EM

Tra

nscript

4S

EM

Tra

nscript

4I

SE

MT

ranscript

4Ia

SE

MT

ranscript

5U

SE

M

116

819

334

3033

611

217

875

130

147

49

778

662

8502

250

7037

662

1047

331

494

352

110

758

3567

997

820

650

626

849

246

297

225

821

926

2695

349

1735

690

165

642

53

196

925

8575

876

10

474

552

2153

050

566

539

98

753

5736

379

853

075

2687

693

405

682

332

797

301

2500

296

1907

844

173

990

42

163

158

7777

091

11

908

521

812

804

642

958

165

042

5725

543

386

033

7064

146

1121

427

428

543

946

957

708

1754

551

355

604

42

009

500

5228

511

11

301

671

809

509

259

673

5232

5503

125

1461

183

2314

294

34

110

535

898

123

2147

628

424

169

116

076

43

299

125

6898

199

12

533

328

846

855

803

913

462

009

6332

481

1153

024

361

475

257

062

632

187

702

5221

964

2060

237

284

715

39

695

102

3683

345

10

208

729

869

931

338

966

24

719

5870

873

1504

872

1002

167

288

996

730

203

203

5233

968

379

946

38

625

45

452

881

4157

836

9601

182

2302

385

349

413

42

170

2948

568

512

800

607

117

305

044

824

701

252

2430

791

937

371

135

651

27

730

762

11

410

958

9272

581

1806

170

1991

100

1322

643

4034

720

748

040

507

186

351

850

9548

283

233

342

00

3085

447

996

042

1354

477

127

313

439

410

120

139

3331

966

1290

930

23

594

23

594

10

20

750

892

6995

886

5414

304

377

157

33

633

921

9572

107

12

496

858

2709

764

340

300

179

715

5324

640

1068

609

3607

137

670

385

11

62

365

62

365

7782

7782

3745

050

676

941

975

521

206

086

217

470

89

376

2478

577

728

930

145

503

77

136

12

10

655

801

1197

429

264

113

52

837

29

318

259

1450

061

4154

594

598

850

450

621

165

116

3801

774

755

829

723

875

325

448

13

17

162

146

1246

170

147

600

7284

40

373

250

7071

001

6962

680

547

429

360

101

133

526

4860

465

583

748

553

968

107

364

14

23

458

544

1360

074

399

089

197

641

42

397

240

4352

339

10

673

156

2033

823

398

685

84

571

6576

002

630

643

971

938

210

600

15

9125

926

1290

662

79

283

17

134

30

494

806

4613

816

8678

579

1109

594

432

938

153

543

3514

849

622

728

347

099

162

303

16

6115

309

886

127

16

400

4481

12

872

250

4527

258

5255

096

1129

854

498

591

78

494

3152

653

1020

838

551

649

303

259

17

28

013

521

674

601

807

373

59

811

29

970

886

4324

265

9351

758

1232

726

624

851

15

928

5789

857

1285

312

377

797

98

235

18

826

278

501

452

103

969

2526

2892

247

933

019

2520

967

1536

839

548

652

690

48

2359

939

996

364

1549

390

346

536

19

19

706

846

1486

934

497

490

69

601

27

396

653

2341

121

5234

801

576

683

484

749

165

279

3608

245

994

093

1780

037

348

233

20

12

867

501

1435

986

142

368

46

546

12

926

583

2557

539

4654

449

2158

951

433

360

99

810

2957

292

239

784

1719

758

505

184

21

5994

249

924

104

55

282

1464

21

411

677

688

809

2224

165

307

796

629

057

377

012

2461

246

853

352

552

120

186

743

22

2253

884

2098

652

68

890

47

082

15

334

332

2734

251

6917

591

3147

661

340

201

41

342

3625

742

135

023

939

412

746

414

23

6357

706

424

651

253

155

53

347

21

239

597

1414

423

2238

072

577

125

453

423

46

699

2328

355

617

905

827

129

244

341

24

2346

569

405

809

99

305

52

009

5601

672

890

313

1475

739

334

767

414

905

62

569

4712

986

637

767

853

815

357

443

Valu

es

liste

dare

pix

eld

ensity

valu

es

±S

EM

.N

um

bers

1th

rough

24

inth

evert

icalc

olu

mn

identif

yth

esam

ple

sused

with

BD

NF

transcripts

and

sta

ndard

err

or

ofth

em

ean

liste

din

the

top

horizonta

l

row

.T

he

pix

elvalu

es

that

are

underlin

ed

are

extr

em

eoutli

ers

.

1060 D. Garzon et al.

� 2002 International Society for Neurochemistry, Journal of Neurochemistry, 82, 1058–1064

Page 4: A new brain-derived neurotrophic factor transcript and decrease inbrain-derived neurotrophic factor transcripts 1, 2 and 3 in Alzheimer's disease parietal cortex

t-tests and Fig. 3. Comparisons of the effects of age and post-

mortem interval on yield of total RNA were done by regression

analysis and two-samples paired t-tests. All statistics were calculated

and results graphed using Microsoft Excel (Microsoft, WA, USA)

and SPSS 10.1 software (SPSS Inc., Chicago, IL, USA).

Results

Samples

The samples consisted of 12 control and 12 AD post-mortem

parietal cortex samples. The average age of the subjects in the

control group was 77.58 ± 2.63 years and that of the AD

group was 78 ± 2.49 years ( p ¼ 0.9). The average post-

mortem delay was 6.23 ± 0.039 h for the control group but

only 3.05 ± 0.028 hours for the AD subjects ( p < 0.001).

However, no significant differences were observed in the yield

of total RNA extracted from both groups (485.62 ± 15.36lg/gof tissue for control and 487.82 ± 29.23 lg/g of tissue for ADsubjects, p ¼ 0.93), or in the integrity of the purified RNA.

Regression analysis yielded no significant correlation between

yield of total RNA and age [r ¼ 0.117 for control (p ¼ 0.71)

and r ¼ 0.382 for AD (p ¼ 0.22)]. Also, regression analysis

for post-mortem delay and yield of total RNA resulted in no

significant correlation [r ¼ 0.312 (p ¼ 0.35) for control and

r ¼ 0.037 (p ¼ 0.91) for AD samples] (data not shown). We

have previously demonstrated no significant correlation

between BDNF mRNA content and age or post-mortem delay

in both control and AD parietal cortex samples (Holsinger

et al. 2000).

New alternative splice site in transcript 4I

The reported size of transcript 4I is 414 bp (Aoyama et al.

2001). Upon PCR amplification, the 414 bp band was faintly

present but was secondary in intensity to a 313-bp band

(Fig. 1). Purification and sequence analysis of the 313-bp

band revealed a new splice variant of exon 4I with splicing

occurring 151 bp from the start of the 5¢ primer and splicingout a 101-bp sequence (Fig. 2).

b-Actin control

To control for variation between samples we used the

constitutively expressed cytoskeletal protein, b-actin. Previ-ous studies from ours and other laboratories have shown no

significant difference in b-actin levels between normal andAD subjects (Takeda et al. 1991; Takeda et al. 1992;

Holsinger et al. 2000). Our results support these previous

findings; statistical comparisons between control and AD

samples yielded no significant difference in b-actin mRNAlevels (Fig. 3, p > 0.05).

Transcripts 1, 2 and 3 are decreased in AD parietal cortex

The two-way ANOVA revealed that overall expression of

transcripts was significantly lower in AD patients compared

Fig. 1 Ethidium bromide-stained gel showing RT-PCR products for

transcript 4I. Lane 1 is the 100 bp DNA ladder. The faint band at

414 bp in lane 2 is the transcript 4I reported by Aoyama et al. (2001).

The intense band at 313 bp in lane 2 is the newly discovered transcript

4Ia. The negative control, RT-PCR without reverse transcriptase, is in

lane 3.

Fig. 2 Sequence of transcript 4Ia. The sequence shown is the human

brain-derived neurotrophic factor transcript 4I (Aoyama et al. 2001).

The underlined sequences at the ends indicate the primers used, and

the middle underlined sequence is the 101 bp region that is spliced out

of transcript 4I, resulting in the new transcript 4Ia. Note: The forward

slash marks the start of the sequence of the mature coding exon 5.

Brain-derived neurotrophic factor transcripts 1061

� 2002 International Society for Neurochemistry, Journal of Neurochemistry, 82, 1058–1064

Page 5: A new brain-derived neurotrophic factor transcript and decrease inbrain-derived neurotrophic factor transcripts 1, 2 and 3 in Alzheimer's disease parietal cortex

with controls [for group, F6,162 ¼ 4.998, p < 0.001)]. This is

consistent with our previous results using the coding exon of

BDNF as the RT-PCR target (Holsinger et al. 2000). Tests of

between-subjects effects revealed a significant difference in

individual transcripts. The difference was statistically signi-

ficant for transcript 1 [F1,22 ¼ 1.018, p ¼ 0.027], transcript 2

[F1,20 ¼ 31.674, p ¼ 0.006] and transcript 3 [F1,20 ¼ 2.115,

p ¼ 0.001], and at the border of statistical significance for

transcript 4 [F1,22 ¼ 1.255, p ¼ 0.062]. None of the other

transcripts [transcript 4I, F1,22 ¼ 3.832, p ¼ 0.456; transcript

4Ia, F1,22 ¼ 0.126, p ¼ 0.202; or transcript 5 U, F1,21 ¼7.077, p ¼ 0.547] demonstrated significant differences

between control and AD (Fig. 3).

Discussion

Using RT-PCR on 24 age- and gender-matched control and

AD samples from the parietal cortex, we report a significant

decrease in three human BDNF mRNA transcripts in the

parietal cortex of AD samples compared with controls. A

two-way ANOVA showed a significant effect for group and

group · transcript. Post-hoc t-tests revealed a significant

difference between control and AD samples for transcript

1, transcript 2 and transcript 3. None of the other transcripts

(4, 4I, 4Ia or 5U) demonstrated any significant differences.

We have previously shown that BDNF mRNA levels are

decreased in the AD parietal cortex compared with controls

(Holsinger et al. 2000). The decreased expression we

demonstrate here in transcripts 1, 2 and 3 in AD could

account for the decreased BDNF expression seen in previous

studies examining the coding exon.

Six non-coding exons and their resulting transcripts have

been reported for the human BDNF gene (exons 1, 2, 3, 4, 4I

and 5U) (Aoyama et al. 2001). Within transcript 4I, we noted

a new splice variant that was more highly expressed in

parietal cortex tissue than the original 414 bp transcript 4I

Fig. 3 Relative levels of mRNA for all brain-

derived neurotrophic factor and b-actin

transcripts in control versus Alzheimer

disease samples. The y-axes show pixel

intensity values determined by phosphorim-

age analysis. Error bars represent standard

error of the mean. Statistically significant

p-values are shown.

1062 D. Garzon et al.

� 2002 International Society for Neurochemistry, Journal of Neurochemistry, 82, 1058–1064

Page 6: A new brain-derived neurotrophic factor transcript and decrease inbrain-derived neurotrophic factor transcripts 1, 2 and 3 in Alzheimer's disease parietal cortex

reported by Aoyama et al. (2001). Sequence analysis

revealed a different splice variant of the 4I transcript (4Ia)

containing a 101-bp deletion.

Although the regulatory elements and factors governing

human BDNF expression are not known, we can draw

parallels with the control of BDNF expression in the rat. In

the rat, promoters I and III are both regulated by calcium

(Tao et al. 1998; Tabuchi et al. 2000). Calcium influx leads

to cAMP response element binding protein (CREB) phos-

phorylation, and phosphorylated CREB binds to and acti-

vates the cAMP response element (CRE) in rat BDNF

promoter III (Shieh et al. 1998; Tao et al. 1998; Shieh and

Ghosh 1999; West et al. 2001). Recently, promoter I was

also reported to be CREB-dependent (Tabuchi et al. 2002).

We have identified consensus CRE sites upstream of exons 1

and 3 in the human BDNF gene which suggests the human

BDNF gene may be regulated in a manner similar to the rat

gene. Levels of phosphorylated CREB are significantly

decreased in post-mortem AD brain samples (Yamamoto

et al. 1999), and a recent study demonstrates that Ab(1–42)lowers CREB phosphorylation, causing decreased expression

of the exon III BDNF transcript in rat cultured cortical

neurons (Tong et al. 2001). Thus, our data implicating down-

regulation of transcripts 1 and 3 in reduced BDNF expression

in AD are consistent with known BDNF regulation in the rat.

On the other hand, the regulatory factors and contribution to

CNS BDNF expression for transcript 2 are still unknown.

In summary, we have shown here that only three of the

seven human BDNF transcripts expressed in brain are down-

regulated in AD. Whether the promoters governing these

transcripts are regulated in a similar manner in the human

CNS and the rat is still unknown. Further investigation will

be necessary to identify the factors that regulate the human

BDNF gene in AD.

Acknowledgements

This work was supported by grants from the Scottish Rite Charitable

Foundation to DG and MF, and from the Ontario Neurotrauma

Foundation to GY and MF. Also a special thanks goes to Dr Henry

Szechtman, McMaster University, and Jennifer Dunn, University of

Toronto, for their assistance with statistical analysis.

References

Alderson R. F., Alterman A. L., Barde Y. A. and Lindsay R. M. (1990)

Brain-derived neurotrophic factor increases survival and differen-

tiated functions of rat septal cholinergic neurons in culture. Neuron

5, 297–306.

Aoyama M., Asai K., Shishikura T., Kawamoto T., Miyachi T., Yokoi T.,

Togari H., Wada Y., Kato T. and Nakagawara A. (2001) Human

neuroblastomas with unfavorable biologies express high levels of

brain-derived neurotrophic factor mRNA and a variety of its

variants. Cancer Lett. 164, 51–60.

Castren E., Pitkanen M., Sirvio J., Parsadanian A., Lindholm D.,

Thoenen H. and Riekkinen P. J. (1993) The induction of LTP

increases BDNF and NGF mRNA but decreases NT-3 mRNA in

the dentate gyrus. Neuroreport 4, 895–898.

Connor B., Young D., Yan Q., Faull R. L., Synek B. and Dragunow M.

(1997) Brain-derived neurotrophic factor is reduced in Alzheimer’s

disease. Mol. Brain Res. 49, 71–81.

Coyle J. T., Price D. L. and DeLong M. R. (1983) Alzheimer’s disease:

a disorder of cortical cholinergic innervation. Science 219,

1184–1190.

Cuello A. C. and Sofroniew M. V. (1984) The anatomy of the CNS

cholinergic neurons. Trends Neurosci. 7, 74–78.

Dragunow M., Beilharz E., Mason B., Lawlor P., Abraham W.

and Gluckman P. (1993) Brain-derived neurotrophic factor

expression after long-term potentiation. Neurosci. Lett. 160, 232–

236.

Ernfors P., Ibanez C. F., Ebendal T., Olson L. and Persson H. (1990)

Molecular cloning and neurotrophic activities of a protein with

structural similarities to nerve growth factor: developmental and

topographical expression in the brain. Proc. Natl Acad. Sci. USA

87, 5454–5458.

Etienne P., Robitaille Y., Wood P., Gauthier S., Nair N. P. and Quirion R.

(1986) Nucleus basalis neuronal loss, neuritic plaques and choline

acetyltransferase activity in advanced Alzheimer’s disease. Neu-

roscience 19, 1279–1291.

Falkenberg T., Mohammed A. K., Henriksson B., Persson H., Winblad

B. and Lindefors N. (1992) Increased expression of brain-derived

neurotrophic factor mRNA in rat hippocampus is associated with

improved spatial memory and enriched environment. Neurosci.

Lett. 138, 153–156.

Ferrer I., Marin C., Rey M. J., Ribalta T., Goutan E., Blanco R., Tolosa

E. and Marti E. (1999) BDNF and full-length and truncated TrkB

expression in Alzheimer disease. Implications in therapeutic

strategies. J. Neuropathol. Exp. Neurol. 58, 729–739.

Ghosh A., Carnahan J. and Greenberg M. E. (1994) Requirement for

BDNF in activity-dependent survival of cortical neurons. Science

263, 1618–1623.

Hefti F. and Weiner W. J. (1986) Nerve growth factor and Alzheimer’s

disease. Ann. Neurol. 20, 275–281.

Hock C., Heese K., Hulette C., Rosenberg C. and Otten U. (2000)

Region-specific neurotrophin imbalances in Alzheimer disease:

decreased levels of brain-derived neurotrophic factor and increased

levels of nerve growth factor in hippocampus and cortical areas.

Arch. Neurol. 57, 846–851.

Hofer M., Pagliusi S. R., Hohn A., Leibrock J. and Barde Y. A. (1990)

Regional distribution of brain-derived neurotrophic factor mRNA

in the adult mouse brain. EMBO J. 9, 2459–2464.

Holsinger R. M., Schnarr J., Henry P., Castelo V. T. and Fahnestock M.

(2000) Quantitation of BDNF mRNA in human parietal cortex by

competitive reverse transcription-polymerase chain reaction:

decreased levels in Alzheimer’s disease. Mol. Brain Res. 76,

347–354.

Hyman C., Hofer M., Barde Y. A., Juhasz M., Yancopoulos G. D.,

Squinto S. P. and Lindsay R. M. (1991) BDNF is a neurotrophic

factor for dopaminergic neurons of the substantia nigra. Nature

350, 230–232.

Kang H. and Schuman E. M. (1995) Long-lasting neurotrophin-induced

enhancement of synaptic transmission in the adult hippocampus.

Science 267, 1658–1662.

Khatchaturian Z. M. (1985) Diagnosis of Alzheimer’s disease. Arch.

Neurol. 42, 1097–1105.

Knusel B., Winslow J. W., Rosenthal A., Burton L. E., Seid D. P.,

Nikolics K. and Hefti F. (1991) Promotion of central choliner-

gic and dopaminergic neuron differentiation by brain-derived

neurotrophic factor but not neurotrophin 3. Proc. Natl Acad. Sci.

USA 88, 961–965.

Brain-derived neurotrophic factor transcripts 1063

� 2002 International Society for Neurochemistry, Journal of Neurochemistry, 82, 1058–1064

Page 7: A new brain-derived neurotrophic factor transcript and decrease inbrain-derived neurotrophic factor transcripts 1, 2 and 3 in Alzheimer's disease parietal cortex

Kokaia Z., Metsis M., Kokaia M., Bengzon J., Elmer E., Smith M. L.,

Timmusk T., Siesjo B. K., Persson H. and Lindvall O. (1994) Brain

insults in rats induce increased expression of the BDNF gene

through differential use of multiple promoters. Eur. J. Neurosci.

6, 587–596.

Lindholm D., Carroll P., Tzimagiogis G. and Thoenen H. (1996)

Autocrine-paracrine regulation of hippocampal neuron survival by

IGF-1 and the neurotrophins BDNF, NT-3 and NT-4. Eur. J.

Neurosci. 8, 1452–1460.

Lowenstein D. H. and Arsenault L. (1996) The effects of growth factors

on the survival and differentiation of cultured dentate gyrus neu-

rons. J. Neurosci. 16, 1759–1769.

Maisonpierre P. C., Le Beau M. M., Espinosa R. III, Ip N. Y., Belluscio

L., de la Monte S. M., Squinto S., Furth M. E. and Yancopoulos

G. D. (1991) Human and rat brain-derived neurotrophic factor and

neurotrophin-3: gene structures, distributions, and chromosomal

localizations. Genomics 10, 558–568.

Mann D. M. (1991) Is the pattern of nerve cell loss in aging and Alz-

heimer’s disease a real, or only an apparent, selectivity? Neurobiol.

Aging 12, 340–343.

McKhann G., Drachman D., Folstein M., Katzman R., Price D. and

Stadlan E. (1984) Clinical Diagnosis of Alzheimer’s Disease.

Report of NINCDS-ADRDA Work Group under the auspices of

Department of Health and Human Service Task Force on Alzhei-

mer’s disease. Neurology 13, 939–944.

McLean B. M., Pittman A. J. and Lo D. C. (2000) Brain-derived neu-

rotrophic factor differentially regulates excitatory and inhibitory

synaptic transmission in hippocampal cultures. J. Neurosci. 20,

3221–3232.

Metsis M., Timmusk T., Arenas E. and Persson H. (1993) Differential

usage of multiple brain-derived neurotrophic factor promoters in

the rat brain following neuronal activation. Proc. Natl Acad. Sci.

USA 90, 8802–8806.

Narisawa-Saito M., Wakabayashi K., Tsuji S., Takahashi H. and Nawa

H. (1996) Regional specificity of alterations in NGF, BDNF and

NT-3 levels in Alzheimer’s disease. Neuroreport 7, 2925–2928.

Osehobo P., Adams B., Sazgar M., Xu Y., Racine R. J. and Fahnestock

M. (1999) Brain-derived neurotrophic factor infusion delays

amygdala and perforant path kindling without affecting

paired-pulse measures of neuronal inhibition in adult rats.

Neuroscience 92, 1367–1375.

Patterson S. L., Grover L. M., Schwartzkroin P. A. and Bothwell M.

(1992) Neurotrophin expression in rat hippocampal slices: a

stimulus paradigm inducing LTP in CA1 evokes increases in

BDNF and NT-3 mRNAs. Neuron 9, 1081–1088.

PhillipsH. S., Hains J.M., LarameeG.R., RosenthalA. andWinslow J.W.

(1990) Widespread expression of BDNF but not NT3 by target

areas of basal forebrain cholinergic neurons. Science 250, 290–

294.

PhillipsH. S., Hains J.M., ArmaniniM., LarameeG. R., Johnson S.A. and

Winslow J. W. (1991) BDNF mRNA is decreased in the hippo-

campus of individualswithAlzheimer’s disease.Neuron 7, 695–702.

Scharfman H. E. (1997) Hyperexcitability in combined entorhinal/hip-

pocampal slices of adult rat after exposure to brain-derived neu-

rotrophic factor. J. Neurophysiol. 78, 1082–1095.

Shieh P. B. and Ghosh A. (1999) Molecular mechanisms underlying

activity-dependent regulation of BDNF expression. J. Neurobiol.

41, 127–134.

Shieh P. B., Hu S. C., Bobb K., Timmusk T. and Ghosh A. (1998)

Identification of a signaling pathway involved in calcium regula-

tion of BDNF expression. Neuron 20, 727–740.

St Amand D., Pottage C., Henry P. and Fahnestock M. (1996) Method

for quantitation of low-abundance nerve growth factor mRNA

expression in human nervous tissue using competitive reverse

transcription polymerase chain reaction. DNA Cell. Biol. 15, 415–

422.

Tabuchi A., Nakaoka R., Amano K., Yukimine M., Andoh T., Ku-

raishi Y. and Tsuda M. (2000) Differential activation of brain-

derived neurotrophic factor gene promoters I and III by Ca2+

signals evoked via 1-type voltage-dependent and N-methyl-

D-aspartate receptor Ca2+ channels. J. Biol. Chem. 275, 17269–

17275.

Tabuchi A. et al. (2002) Involvement of an upstream stimulatory factor

as well as cAMP responsive element-binding protein in the acti-

vation of brain-derived neurotrophic factor gene promoter I.

J. Biol. Chem., in press.

Takeda M., Nishimura T., Hariguchi S., Tatebayashi Y., Tanaka T.,

Tanimukai S. and Tada K. (1991) Study of cytoskeletal proteins in

fibroblasts cultured from familial Alzheimer’s disease. Acta Neu-

rol. Scand. 84, 416–420.

Takeda M., Tatebayashi Y. and Nishimura T. (1992) Change in the

cytoskeletal system in fibroblasts from patients with familial

Alzheimer’s disease. Prog. Neuropsychopharmacol. Biol.

Psychiatry 16, 317–328.

Tao X., Finkbeiner S., Arnold D. B., Shaywitz A. J. and Greenberg M. E.

(1998) Ca2+ influx regulates BDNF transcription by a CREB

family transcription factor-dependent mechanism. Neuron 20, 709–

726.

Timmusk T., Palm K., Metsis M., Reintam T., Paalme V., Saarma M. and

Persson H. (1993) Multiple promoters direct tissue-specific

expression of the rat BDNF gene. Neuron 10, 475–489.

Timmusk T., Lendahl U., Funakoshi H., Arenas E., Persson H. and

Metsis M. (1995) Identification of brain-derived neurotrophic

factor promoter regions mediating tissue-specific, axotomy-, and

neuronal activity-induced expression in transgenic mice. J. Cell.

Biol. 128, 185–199.

Tong L., Thornton P. L., Balazs R. and Cotman C. W. (2001) Beta-

amyloid-(1–42) impairs activity-dependent cAMP-response ele-

ment-binding protein signaling in neurons at concentrations in

which cell survival is not compromised. J. Biol. Chem. 276,

17301–17306.

West A. E., ChenW. G., Dalva M. B., Dolmetsch R. E., Kornhauser J. M.,

Shaywitz A. J., Takasu M. A., Tao X. and Greenberg M. E. (2001)

Calcium regulation of neuronal gene expression. Proc. Natl Acad.

Sci. USA 98, 11024–11031.

Wetmore C., Ernfors P., Persson H. and Olson L. (1990) Localization of

brain-derived neurotrophic factor mRNA to neurons in the brain by

in situ hybridization. Exp. Neurol. 109, 141–152.

Whitehouse P. J., Price D. L., Struble R. G., Clark A. W., Coyle J. T.

and Delon M. R. (1982) Alzheimer’s disease and senile

dementia: loss of neurons in the basal forebrain. Science 215,

1237–1239.

Yamamoto M., Ozawa H., Saito T., Rosler M. and Riederer P. (1999)

Impaired phosphorylation of cyclic AMP response element binding

protein in the hippocampus of dementia of the Alzheimer type.

Brain Res. 824, 300–303.

1064 D. Garzon et al.

� 2002 International Society for Neurochemistry, Journal of Neurochemistry, 82, 1058–1064