40
1 p300 Acetyltransferase Activity Differentially Regulates the 1 Localization and Activity of the FOXO Homologues in Skeletal 2 Muscle 3 Sarah M. Senf 1 , Pooja B. Sandesara 2 , Sarah A. Reed 2 , Andrew R. Judge 1,2 4 1Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida, 5 USA; 2 Department of Physical Therapy, University of Florida, Gainesville, Florida, USA 6 7 Corresponding author: 8 Andrew R. Judge, 9 Department of Physical Therapy 10 J303, Biomedical Sciences Building 11 1275 Center Drive 12 University of Florida, 13 Gainesville, FL 32610 14 USA 15 Tel: 352-273-9220 16 Fax: 352-273-6109 17 E-mail: [email protected] 18 19 20 21 22 23 24 Running title: p300 HAT activity represses FOXO in muscle 25 Articles in PresS. Am J Physiol Cell Physiol (March 9, 2011). doi:10.1152/ajpcell.00255.2010 Copyright © 2011 by the American Physiological Society.

p300 Acetyltransferase activity differentially regulates the localization and activity of the FOXO homologues in skeletal muscle

Embed Size (px)

Citation preview

1

p300 Acetyltransferase Activity Differentially Regulates the 1

Localization and Activity of the FOXO Homologues in Skeletal 2

Muscle 3

Sarah M. Senf1, Pooja B. Sandesara2, Sarah A. Reed2, Andrew R. Judge1,2 4

1Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida, 5

USA; 2 Department of Physical Therapy, University of Florida, Gainesville, Florida, USA 6

7

Corresponding author: 8

Andrew R. Judge, 9

Department of Physical Therapy 10

J303, Biomedical Sciences Building 11

1275 Center Drive 12

University of Florida, 13

Gainesville, FL 32610 14

USA 15

Tel: 352-273-9220 16

Fax: 352-273-6109 17

E-mail: [email protected] 18

19

20

21

22

23

24

Running title: p300 HAT activity represses FOXO in muscle 25

Articles in PresS. Am J Physiol Cell Physiol (March 9, 2011). doi:10.1152/ajpcell.00255.2010

Copyright © 2011 by the American Physiological Society.

2

ABSTRACT 26

The Forkhead Box O (FOXO) transcription factors regulate diverse cellular processes, and in 27

skeletal muscle are both necessary and sufficient for muscle atrophy. Although the regulation of 28

FOXO by Akt is well evidenced in skeletal muscle, the current study demonstrates that FOXO is 29

also regulated in muscle via the acetyltransferase (HAT) activities of p300/CBP. Transfection of 30

rat soleus muscle with a dominant-negative (d.n.) p300, which lacks HAT activity and inhibits 31

endogenous p300 HAT activity, increased FOXO reporter activity and induced transcription 32

from the promoter of a bona fide FOXO target gene, atrogin-1. Conversely, increased HAT 33

activity via transfection of either WT p300 or WT CBP repressed FOXO activation in vivo in 34

response to muscle disuse, and in C2C12 cells in response to dexamethasone & acute starvation. 35

Importantly, manipulation of HAT activity differentially regulated the expression of various 36

FOXO target-genes. Co-transfection of FOXO1, FOXO3a or FOXO4 with the p300 constructs 37

further identified p300 HAT activity to also differentially regulate the activity of the FOXO 38

homologues. Markedly, decreased HAT activity strongly increased FOXO3a transcriptional 39

activity, while increased HAT activity repressed FOXO3a activity and prevented its nuclear 40

localization in response to nutrient deprivation. In contrast, p300 increased FOXO1 nuclear 41

localization. In summary, this study provides the first evidence to support the acetyltransferase 42

activities of p300/CBP in regulating FOXO signaling in skeletal muscle, and suggests that 43

acetylation may be an important mechanism to differentially regulate the FOXO homologues and 44

dictate which FOXO target-genes are activated in response to varying atrophic stimuli. 45

46

Key Words: HAT, muscle atrophy, disuse, cachexia, gene regulation, atrogin-1 47

48

3

INTRODUCTION 49

FOXO signaling has been implicated in skeletal muscle atrophy associated with sepsis 50

(9), starvation (24, 27), diabetes (28), cancer (27), aging (16, 27) and heart failure (44). More 51

direct work, using genetic approaches, demonstrate that at least two of the FOXO homologues, 52

FOXO1 (23) and FOXO3a (43), are sufficient to cause skeletal muscle atrophy, in vivo. Perhaps 53

more importantly, blocking FOXO transactivation prevents at least 40% of disuse muscle fiber 54

atrophy (39, 45), further demonstrating the requirement of FOXO for the normal atrophy 55

phenotype in a physiological model of muscle atrophy. Given the significance of FOXO in the 56

regulation of muscle mass, identifying the immediate upstream regulators of FOXO may lead to 57

the development of specific countermeasures to prevent muscle wasting. 58

The regulation of FOXO signaling by Akt has been extensively characterized in a variety 59

of cell types, including skeletal muscle (2, 26, 43). In response to growth conditions or growth 60

factor stimuli the IGF-1/PI3K/Akt pathway is activated, which leads to FOXO phosphorylation 61

by Akt on specific residues, which promotes the cytosolic retention and inactivation of FOXO (2, 62

42). In skeletal muscle, direct evidence to support the IGF-1/PI3K/Akt pathway in regulating 63

FOXO can be found in several studies (26, 43, 48). Decreases in this signaling pathway in 64

skeletal muscle during physiological conditions of muscle atrophy such as starvation and muscle 65

disuse are thought to contribute to FOXO activation. However, increasing evidence demonstrate 66

FOXO-signaling to be controlled via additional post-translational modifications and protein-67

protein interactions which are distinct from Akt-mediated phosphorylation (19, 51). Yet, many of 68

these additional regulatory mechanisms have yet to be thoroughly explored in skeletal muscle. If 69

similar control mechanisms indeed exist in skeletal muscle to modulate FOXO activity, this 70

4

could potentially open up new avenues for therapeutically blocking FOXO function and the 71

associated muscle atrophy during physiological conditions of muscle wasting. 72

One such mechanism of FOXO regulation identified in multiple cell types, involves the 73

regulation of FOXO-dependent transcription by the histone acetyltransferase (HAT) proteins, 74

p300 and CREB binding protein (CBP) (11, 14, 35). These HAT proteins each possess an 75

intrinsic acetyltransferase activity which catalyzes the transfer of an acetyl group to specific 76

lysine residues on target proteins (10, 11, 32). Although HATs are most well known for 77

regulating gene transcription through histone acetylation and relaxation of chromatin structure at 78

gene promoters (12, 31), HATs also play an important role in regulating the activity of a variety 79

of transcription factors, including p53, MyoD, HIF-1alpha, as well as the FOXO transcription 80

factors (47, 52). HATs may regulate transcription factor activity through various mechanisms 81

which include interaction and recruitment of factors to target gene promoters, via acting as 82

adaptor molecules facilitating protein-protein interactions, and through direct acetylation of 83

transcription factors or other necessary co-factors which thereby alter transcription factor activity 84

(25). Evidence for HAT-mediated regulation of the FOXO transcription factors can be found in 85

multiple cell types. Interestingly however, the resulting effect of HATs on FOXO appears to be 86

cell-type specific and/or specific to the FOXO homologue. For example, p300 increases 87

FOXO1-dependent transcription from the IGFBP-1 promoter reporter in H4IIE rat hepatoma 88

cells, which requires p300 HAT activity (35). Similarly, p300 increases FOXO3a-dependent 89

transcription from the Bim promoter in human embryonic kindey cells (HEK293T) (34). In 90

contrast, p300 represses FOXO4-induced transcription of GADD45, p27, p21 and MnSOD in 91

HEK293 cells (11). Therefore depending on the cell type, FOXO homologue, and target gene 92

measured, HATs may either repress or activate FOXO-induced transcription, which may reflect 93

5

an important fine-tuning mechanism of FOXO-target gene regulation. However, despite the 94

importance of understanding the mechanisms which lead to FOXO-dependent transcription in 95

skeletal muscle due to its known role in causing muscle atrophy, no data currently exist to 96

suggest whether the acetyltransferase activities of p300/CBP regulate FOXO in skeletal muscle. 97

Therefore, the purpose of the current study was to determine whether HAT proteins regulate the 98

FOXO transcription factors in skeletal muscle, and whether this is altered during conditions of 99

muscle wasting. 100

101

102

103

104

105

106

107

108

109

110

111

112

113

114

115

116

6

MATERIALS AND METHODS 117

Animals 118

Sprague-Dawley male rats (200-225g) were ordered from Charles River Laboratories 119

(Wilmington, MA, USA). The University of Florida Institutional Animal Care and Use 120

Committee approved all animal procedures. UF is accredited by the Association for Assessment 121

and Accreditation of Laboratory Animal Care (#A3377-01). Animals were maintained in a 122

temperature and humidity-controlled facility with a 12-hour light/dark cycle. Water and standard 123

diet were provided ad libitum. 124

Plasmids and Reporter Gene Assays 125

Expression plasmids for WT p300 and the dominant negative (d.n.) p300 mutant (which lacks 126

acetyltransferase activity due to an inactivating point mutation, converting aspartic acid 1399 to 127

tyrosine ) were obtained from Dr. Tso-Pang Yao (Duke University, Durham, NC, USA) and have 128

been previously described (21). The FOXO1 expression plasmid was a gift from Dr. Akiyoshi 129

Fukamizu (University of Tsukuba, Ibaraki, Japan) and has previously been used and described 130

(32). The tagged FOXO1-EGFP plasmid was obtained from Addgene (plasmid 9022), and was 131

deposited by Dr. Domenico Accili (Columbia University, New York, NY, USA) and has 132

previously been described (13). The FOXO4 expression plasmid was a gift from Dr. Boudewijn 133

Burgering (University Medical Center, Utrecht, The Netherlands) and has been previously used 134

and described (53). The FOXO3a expression plasmid was obtained from Addgene (plasmid 135

10710), was deposited by Dr. William Sellers (Novartis, Cambridge, MA) and has been 136

previously used and described (37). The WT FOXO3a-DsRed fusion construct was created via 137

PCR amplification of the FOXO3a cDNA out of the parent vector using primers to create 138

HindIII and SalI restriction sites on the 5’ and 3’ ends of the FOXO3a coding region, 139

7

respectively. FOXO3a cDNA was then sub-cloned, in frame, into the DsRed2-c1 plasmid. 140

Verification that FOXO3a cDNA was in frame was confirmed via DNA sequencing (DNA 141

Sequencing Core, University of Florida). The d.n.Akt and c.a.Akt expression plasmids were 142

obtained from Addgene (plasmids 12643 & 16244, respectively) and were deposited by Dr. 143

Mien-Chie Hung (The University of Texas, M. D. Anderson Cancer Center, Houston, TX) and 144

have previously been described (58). The DAF-16/FOXO responsive reporter plasmid, the 145

atrogin-1-GL2 promoter reporter plasmid and the d.n.FOXO construct have also been previously 146

used and described (46). pRL-TK-Renilla was purchased from Promega. Plasmid DNA was 147

amplified and isolated from bacterial cultures using Endotoxin-Free Maxi or Mega Prep Kits 148

(Qiagen, Valencia, CA, USA), precipitated in ethanol and re-suspended in 1X sterile filtered 149

phosphate buffered saline (PBS) for in vivo transfections, or Tris-EDTA (TE) buffer for 150

transfections in culture. 151

In vivo, Plasmid Injection and Electroporation. 152

Transfection of plasmid DNA into skeletal muscle, in vivo, has been detailed previously (22, 46). 153

For rat experiments, 10μg each of the expression or control plasmid(s) and 40μg of the reporter 154

plasmid were diluted in a total of 50μl 1XPBS for each solei injection. Standard procedures 155

were used to determine luciferase activity on skeletal muscle homogenates using a Modulus 156

single tube multimode reader (Promega) and have been described previously (46). 157

Animal Models and Muscle Preparation 158

Disuse muscle atrophy via cast immobilization of both hind limbs was induced in rats four days 159

following plasmid injection and has been detailed previously (46). After three days of 160

immobilization or weight bearing activity soleus muscles were removed and processed either 161

immediately for RNA isolation or frozen in liquid nitrogen and stored at -80°C until further 162

8

biochemical analyses. For experiments using exclusively genetic manipulations, muscles were 163

harvested 7 days post-plasmid injection. 164

Cell Culture Experiments 165

C2C12 cells were cultured on 0.1% gelatin coated 6-well plates in high-glucose DMEM 166

(Invitrogen), 10% fetal bovine serum and 5% CO2. Muscle cells were transfected with plasmid 167

DNA at ~80% confluence using FuGENE® HD Transfection Reagent (Promega Corp, Madison, 168

WI, USA) at a 3.5:1 ratio of reagent to total DNA. Sixteen hours following transfection muscle 169

cells were differentiated into myotubes by incubation in differentiation medium (2% Horse 170

Serum in DMEM). For dexamethasone studies, 6-day differentiated myotubes were treated with 171

either vehicle (water) or 1uM water-soluble dexamethasone (Sigma, St. Louis, MO, USA) in 172

differentiation media for 6 hours and harvested in Passive Lysis Buffer (Promega). In the 173

nutrient deprivation groups, differentiation media was removed from 4-day differentiated cells 174

and Hanks Balanced Salt Solution (HBSS) added for either 2 hours (localization experiments) or 175

6 hours (reporter assays and gene expression) prior to harvest. To inhibit PI3Kinase, 10μM 176

LY294002 (Calbiochem, Merck KGaA, Darmstadt, Germany) or vehicle (ethanol) was added to 177

4-day differentiated myotubes for 6 hours. For reporter experiments, cells were harvested in 178

Passive Lysis Buffer (Promega) and luciferase activity determined by normalizing firefly 179

luciferase activity to pRL-TK Renilla luciferase activity using a Dual-Luciferase® Reporter 180

Assay (Promega). 181

RNA isolation, cDNA synthesis, and RT-PCR 182

RNA isolation and cDNA synthesis from whole muscle was performed using a Trizol-based 183

method as previously described (46). RNA isolation from C2C12 myotubes was performed 184

similarly, following addition of 250uL Trizol per well and vigorous scraping, as previously 185

9

described (33) . cDNA was generated from 1ug of RNA and was used as a template for qRT-186

PCR using primers for atrogin-1, GenBank NM_133521; MuRF1, GenBank NM_080903; 187

Cathepsin-L, GenBank NM_013156; 4E-BP1, GenBank NM_053857; LC3b, GenBank 188

NM_022867; p21, Genbank NM_080782; Gadd45α, GenBank NM_024127; Foxo1, GenBank 189

NM_001191846; Foxo3a, GenBank NM_001106395; Foxo4, GenBank NM_001106943; or 18S, 190

GenBank X03205.1, which were ordered from Applied Biosystems (Austin, TX, USA). TaqMan 191

probe-based chemistry was used to allow detection of PCR products using a 7300 real-time PCR 192

system (Applied Biosystems), and quantification of gene expression was performed using the 193

relative standard curve method. 194

Western Blotting and Co-Immunoprecipitation Assays 195

Preparation of muscle homogenates and western blotting were performed according to standard 196

procedures and have been described previously (46). Primary antibodies for p300 (#554215, BD 197

Pharmingen, San Jose, CA, USA), FOXO1 (#9454S, Cell Signaling Technology, Boston, MA, 198

USA); phospho-FOXO1 (Ser256) (#9461, Cell Signaling Technology); FOXO3a (SC-11351, 199

Santa Cruz Biotechnology, Santa Cruz, CA, USA); phospho-FOXO3a (Thr32) (SC-12357, Santa 200

Cruz Biotechnology) and FOXO4 (07-1720, Millipore, Billerica, MA), were used according to 201

manufacturer’s directions. Tubulin primary antibody, (T6074 from Sigma-Aldrich Inc, St. 202

Louis, MO, USA) was used to control for equal protein loading and protein transfer. For co-203

immunoprecipitation assays proteins, 500ug of muscle protein were incubated overnight with 204

either 4ug of Anti-Acetyl Lysine antibody (#05-515) or 4ug of anti-p300 (#05-257) using a 205

Catch and Release Reversible Immunoprecipitation System (#17-500), all from Millipore. The 206

following day, precipitated proteins were washed and subsequently eluted in denaturing buffer, 207

boiled, and western blotting performed for endogenous FOXO3a and FOXO1. 208

10

Fluorescent Microscropy 209

C2C12 myoblasts were seeded on 6-well plates containing 0.1% gelatin-coated glass coverslips, 210

transfected and differentiated for 4 days. Following treatment, cells were rinsed with PBS and 211

fixed for 30 minutes in 4% paraformaldehyde. Following three washes in PBS, two drops of 212

Vectashield Mounting Medium for Fluorescence, with Dapi, (#H-1200, Vector Laboratories, Inc, 213

Burlingame, CA, USA) was added to each coverslip. A Leica DM5000B microscope (Leica 214

Microsystems Inc., Bannockburn, IL USA) containing GFP (green) and Rhodamine (red) filter 215

cubes was used to visualize FOXO1-EGFP or FOXO3a-DsRed positive myotubes, respectively. 216

A Dapi (blue) filter was used to visualize Dapi-stained nuclei. Images were captured and merged 217

using Leica Application Suite, version 3.5.0. 218

Statistics 219

Data were analyzed using a two-way ANOVA followed by Bonferroni post-hoc comparisons 220

when appropriate (GraphPad Software, San Diego, CA). All data are expressed as the mean ± 221

SEM, and significance was set at P<0.05. 222

223

224

225

226

227

228

229

230

231

11

RESULTS 232

p300 acetyltransferase activity is necessary and sufficient to repress FOXO signaling in 233

skeletal muscle 234

To determine if p300 HAT activity regulates FOXO signaling in skeletal muscle, we injected and 235

electrotransfered a FOXO-responsive luciferase reporter plasmid plus either a control plasmid, 236

WT p300 or dominant negative (d.n.) p300 (which lacks acetyltransferase activity) expression 237

plasmid into the skeletal muscle of rats prior to either normal weight bearing activity or three 238

days of muscle disuse induced via hind limb cast immobilization. As shown in Figure 1A, 239

FOXO activity was increased (~3-fold) in response to immobilization, which is in agreement 240

with our previous findings (46). However, in muscles overexpressing WT p300, FOXO reporter 241

activation in response to immobilization was prevented. Further, expression of the d.n.p300 242

mutant, which outcompetes and inhibits endogenous p300 HAT activity, was sufficient to 243

increase FOXO activity in normal weight bearing muscle (~6-fold) and further enhanced FOXO 244

activity during immobilization (~14-fold). To determine whether another protein which 245

possesses HAT activity and is highly homologous to p300, CREB-binding protein (CBP), also 246

inhibits disuse-induced activation of the FOXO reporter we collected additional data using a 247

control or WT CBP expression plasmid. Similar to WT p300, overexpression of WT CBP 248

prevented the increase in FOXO activity during disuse (Figure 1B). 249

To test whether p300 and CBP could similarly prevent the activation of FOXO in 250

response to a stimulus distinct from muscle disuse we determined the effect of p300/CBP on 251

FOXO activation, in vitro, in response to treatment with the glucocorticoid, dexamethasone 252

(Dex). C2C12 myoblasts were transfected with the FOXO reporter and pRL-TK-renilla plus an 253

empty vector, WT p300 or WT CBP plasmid. Cells were differentiated for 6 days, and treated 254

12

with either vehicle (water) or Dex, at a concentration of 1µM for 6 hrs. Dex treatment decreases 255

phospho-FOXO protein levels in C2C12 myotubes (43), which is widely used as a marker of 256

FOXO activation and, indeed, we found Dex increased FOXO reporter activity 1.7-fold over 257

control. However, this increase in FOXO activity due to Dex treatment was abolished in C2C12 258

cells transfected with either WT p300 or WT CBP (Figure 1C). These findings provide the first 259

evidence that FOXO activity is regulated in skeletal muscle via the acetyltransferase activity of 260

p300/CBP and further indicate that HAT activity is both necessary and sufficient to repress 261

FOXO activity in skeletal muscle. 262

263

p300 HAT activity differentially regulates FOXO target gene transcription 264

Due to the pronounced effect that p300 HAT activity has on FOXO transcriptional activity in 265

skeletal muscle, we next determined whether p300 could also block the increased transcription of 266

a bona fide FOXO target gene, atrogin-1/MAFbx, during 3 days of muscle disuse. Atrogin-1 267

mRNA was increased 4.7-fold in response to muscle disuse, which was attenuated by 65% in 268

muscles overexpressing WT p300 (Figure 2A). This repression by p300 required its HAT 269

activity since expression of d.n.p300 did not similarly repress atrogin-1 mRNA levels. To further 270

confirm that p300 can repress atrogin-1 transcription during muscle disuse we conducted similar 271

experiments to test the effect of WT p300 or d.n.p300 on a luciferase reporter construct driven by 272

2.4 kb of the atrogin-1 promoter. As shown in Figure 2B, similar to the affect of p300 on 273

atrogin-1 mRNA, WT p300 prevented disuse-induced activation of the atrogin-1 promoter 274

reporter, which required its HAT activity. Notably, the effects of p300 HAT activity were more 275

pronounced on the atrogin-1 promoter reporter when compared to atrogin-1 mRNA levels. This 276

finding is likely explained by the muscle fiber transfection efficiency in whole muscle with the 277

13

p300 constructs, which in our hands is ~50%. Therefore, the effects of WT and d.n.p300 on 278

mRNA levels in transfected fibers are diluted by mRNA levels from non-transfected fibers. In 279

contrast, when co-injecting two plasmids (p300 and the atrogin-1 promoter reporter constructs), 280

co-transfection of a muscle fiber occurs nearly 100% of the time (38). As a result, the atrogin-1 281

promoter reporter is only reporting from those fibers which also took up the p300 (or empty 282

vector) constructs—thereby eliminating the dilution effect. 283

Since d.n.p300 was sufficient to increase both the FOXO reporter and the atrogin-1 284

promoter reporter during normal weight bearing conditions, we further determined whether the 285

increase in atrogin-1 promoter activity by d.n.p300 required active FOXO. Co-expression of 286

d.n.p300 with a d.n.FOXO construct (which inhibits FOXO activity (46)), blocked the ability of 287

d.n.p300 to activate the atrogin-1 promoter reporter (Figure 2C). Collectively these data 288

demonstrate that p300 HAT activity represses atrogin-1 transcription, which is mediated through 289

the repression of FOXO. 290

We next examined the effect of p300 HAT activity on the mRNA levels of additional 291

FOXO target-genes during normal weight bearing conditions and 3 days of immobilization. As 292

shown in Figure 2D, and in agreement with others (1, 4, 22, 30, 57), 3 days of muscle disuse 293

induced a significant increase in the mRNA levels of MuRF1 (3.2-fold), p21 (3.9-fold), 4E-BP1 294

(2-fold), Gadd45a (1.7-fold), cathepsin L (2.4-fold) and LC3b (2.1-fold). However, increasing 295

HAT activity via overexpression of WT p300 significantly repressed the disuse-induced increase 296

in MuRF1 by 55%, and the bona fide FOXO target-gene, p21, by 54% both of which required 297

p300’s HAT activity. In contrast, WT p300 further enhanced the disuse-induced increases in 4E-298

BP1 by 48% and another bona fide FOXO target-gene, Gadd45α, by 75% which required p300’s 299

HAT activity. Importantly, reducing HAT activity via expression of d.n.p300 significantly 300

14

repressed the disuse-induced increases in cathepsin L by 78%, and 4E-BP1 by 45%. Alterations 301

in p300 HAT activity did not significantly affect the levels of LC3b mRNA. Three days of 302

muscle disuse also significantly increased the mRNA levels of FOXO1 (2.2-fold) and FOXO3 303

(1.9-fold), which has previously been demonstrated (40, 45) and also increased the mRNA levels 304

of FOXO4 (2.3-fold) (Figure 2E). Notably, increased p300 HAT activity had a significant 305

repressive effect on FOXO4 mRNA levels during both weight bearing and immobilization 306

conditions and on FOXO1 mRNA levels during immobilization. There was no significant effect 307

of p300 HAT activity on FOXO3a mRNA levels. Together, these data indicate that p300 HAT 308

activity differentially regulates the expression of FOXO target genes, as well as the FOXO genes 309

themselves. 310

311

p300 acetylates and represses FOXO3a transcriptional activity 312

Data thus far demonstrate that p300 HAT activity represses the increase in FOXO transcriptional 313

activity in response to two different atrophic stimuli and differentially regulates the gene 314

expression of FOXO-target genes. Because the FOXO reporter may respond to each of the 315

skeletal muscle FOXO family members, the regulation of total FOXO activity and target gene 316

expression by p300 may be related to the regulation of FOXO1, FOXO3a, FOXO4, or some 317

combination of FOXO factors. We therefore tested the extent to which p300 HAT activity 318

regulates the transcriptional activity of FOXOs 1, 3a or 4 in skeletal muscle. To do this we 319

injected and electrotransfered rat solei with the FOXO reporter plasmid plus FOXO1, FOXO3a 320

or FOXO4 expression plasmids (Figure 3A), each with an empty vector, WT p300, or d.n.p300. 321

As shown earlier in Figure 1A, transfection of WT p300 alone had no affect on basal levels of 322

the FOXO reporter, while transfection of d.n.p300 was sufficient to increase FOXO activity 4.6-323

15

fold. Transfection of FOXO1 alone did not significantly increase the FOXO reporter and did 324

not significantly alter reporter activity in the presence of WT p300 or d.n.p300 (Figure 3B). In 325

contrast, the 2.5-fold increases in FOXO reporter activity induced individually by FOXO3a and 326

FOXO4 were abolished by WT p300. Moreover, the increases in FOXO reporter activity 327

induced individually by FOXO3a (2.5-fold) and d.n.p300 (4.6-fold), were synergistically 328

increased to 16-fold when FOXO3a and d.n.p300 were co-expressed together. Co-expression of 329

FOXO4 plus d.n.p300 resulted in a 7-fold increase in FOXO activity, demonstrating an additive 330

effect on FOXO activity. Together these findings show that p300 is sufficient to repress both 331

FOXO3a and FOXO4 activity via its HAT activity, and suggests that a decrease in p300 332

acetyltransferase activity potently increases the transcriptional activity of FOXO3a. 333

Due to the pronounced effect that p300 HAT activity has on repressing FOXO3a 334

transcriptional activity, we determined whether we could detect changes in endogenous FOXO3a 335

acetylation in muscles injected with the WT p300 and d.n.p300 constructs. Total acetylated 336

proteins were immunoprecipitated from equal amounts of protein extract using an anti-acetyl-337

lysine antibody and subsequently immunoblotted for FOXO3a. As shown in Figure 3C, muscles 338

injected with WT p300 demonstrated an increase in acetylated FOXO3a when compared to 339

muscles injected with an empty vector, while d.n.p300-injected muscles showed a decrease in 340

acetylated FOXO3a. These findings show that FOXO3a is a target of p300 HAT activity and 341

suggest that p300 may regulate FOXO3a transcriptional activity through direct acetylation. 342

Protein modification via acetylation has previously been demonstrated to regulate protein 343

stability (41). Therefore, we determined whether p300 HAT activity regulates FOXO3a protein 344

levels. Due to the relatively low levels of endogenous FOXO3a protein in skeletal muscle and 345

the difficulty that this poses in quantifying a p300-effect on FOXO3a, we measured the effect of 346

16

either WT or d.n.p300 on ectopically expressed FOXO3a. As shown in Figure 3D, co-347

transfection of WT p300 significantly reduced FOXO3a protein, which required its HAT 348

activity, since co-tranfection of d.n.p300 did not similarly reduce FOXO3a protein levels. 349

Although the decrease in FOXO3a protein by WT p300 may explain our finding that WT p300 350

repressed FOXO3a-induced transcription of the FOXO reporter (as shown in Figure 3B), the 351

levels of FOXO3a protein in muscles co-transfected with d.n.p300 were not significantly 352

increased. Therefore, the synergistic increase in FOXO3a-induced reporter activity when HAT 353

activity was decreased (by d.n.p300) cannot be fully accounted for by higher levels of FOXO3a 354

protein. To demonstrate this we normalized FOXO reporter activity from Figure 3B (underlined 355

groups) to total FOXO3a protein levels. Data are normalized such that the ratio of FOXO 356

activity/total FOXO3a protein levels in muscles transfected with FOXO3a alone is set to baseline 357

(Figure 3E). Since co-transfection of WT p300 with FOXO3a blocked FOXO3a-induced 358

reporter activation and concomitantly decreased total FOXO3a levels, the ratio of FOXO 359

activity/total FOXO3a protein did not change in the presence of WT p300. However, the 16-fold 360

increase in FOXO activity in muscles transfected with FOXO3a and d.n.p300, was still increased 361

by more than 4-fold when normalized to total FOXO3a protein levels (Figure 3E). Therefore, 362

regulation of FOXO3a protein levels by p300 HAT activity cannot fully account for the effect of 363

p300 on FOXO3a transcriptional activity. In summary these data demonstrate that p300 HAT 364

activity increases FOXO3a acetylation, and represses the transcriptional capacity of FOXO3a. 365

366

p300 & CBP differentially regulate FOXO1 and FOXO3a cellular localization 367

One mechanism whereby acetylation has been shown to regulate FOXO transcriptional activity 368

is through the regulation of FOXO cellular localization (13, 36). To determine whether 369

17

p300/CBP regulate FOXO cellular localization we transfected C2C12 cells with a FOXO3a-370

DsRed fusion construct plus an empty vector, WT p300 or WT CBP expression plasmid. 371

Following differentiation into myotubes we induced FOXO3a-DsRed nuclear localization via 372

nutrient deprivation for 2 hours, which has been shown previously (7, 43), and determined the 373

effect of p300 and CBP on FOXO3a-DsRed cellular localization via fluorescent microscopy 374

using a Rhodamine (red) filter. The redistribution of FOXO3a-DsRed from the cytosol to the 375

nucleus in response to nutrient deprivation (Figure 4A, B) was confirmed via Dapi staining 376

(blue), and was prevented by p300 (Figure 4D) and attenuated by CBP (Figure 4F). Similarly, 377

during control conditions, FOXO3a-DsRed was more predominantly cytosolic in myotubes co-378

expressing p300 (Figure 4C) or CBP (Figure 4E). Since FOXO1 signaling is also increased in 379

response to nutrient deprivation (15) we further tested the ability of p300 and CBP to regulate 380

FOXO1 cellular localization, using the same experimental design just described, but replacing 381

FOXO3a-DsRed with FOXO1-GFP, and using a GFP (green) filter. Two hours of nutrient 382

deprivation induced a modest, but visible increase in FOXO1-GFP nuclear localization (Figure 383

4G, H), which was potentiated by WT p300 (Figure 4J) and attenuated by WT CBP (Figure 4L). 384

Though less visible, WT p300 similarly increased FOXO1-GFP nuclear localization during 385

control conditions. This effect of p300 on FOXO1-GFP localization in response to starvation is 386

in stark contrast to its effect on FOXO3a-DsRed, further indicating that p300 differentially 387

regulates the FOXO homologues. 388

Since both p300 and CBP were found to regulate FOXO1 and FOXO3a cellular 389

localization, we further measured their ability to regulate total FOXO reporter activation in 390

response to nutrient deprivation. To do this we transfected C2C12 cells with the FOXO reporter 391

plasmid plus an empty vector, WT p300 or WT CBP plasmid. Following four days of 392

18

differentiation, cells either remained in differentiation media (control conditions) or were 393

nutrient deprived by removing the media and incubating in HBSS for 6 hours. FOXO-dependent 394

luciferase activity was increased by ~20% following nutrient deprivation, and was not altered by 395

p300 during either condition (Figure 4M). In contrast, transfection of CBP reduced FOXO-396

dependent luciferase activity during both control and nutrient deprivation conditions when 397

compared to empty vector groups. To determine the physiological relevance of this decrease in 398

total FOXO activity by CBP, we measured the ability of CBP to regulate the gene transcription 399

of the atrophy related FOXO target gene, atrogin-1, following 6 hours of nutrient deprivation. 400

As shown in Figure 4N, 6 hours of HBSS treatment induced a 2.5-fold increase in atrogin-1 401

transcription, which was attenuated by ~60% in HBSS treated cells transfected with CBP. 402

Therefore overexpression of CBP can attenuate both FOXO3a and FOXO1 nuclear localization 403

in response to nutrient deprivation and prevent the full activation of atrogin-1. 404

Our findings in Figure 3B using the FOXO1 and p300 constructs suggest that alterations 405

in p300 HAT activity are not sufficient to regulate FOXO1 transcriptional activity (on the FOXO 406

reporter), in vivo. However, in our localization experiments shown in Figure 4I and 4J, p300 407

strongly induced FOXO1 nuclear localization. Since p300 is predominately localized to the 408

nucleus, we tested the extent to which FOXO1 and p300 interact, which may potentially explain 409

the increased nuclear residence of FOXO1 in p300-transfected cells. Since p300 also regulated 410

FOXO3a localization, though in an opposite manner, we also measured the extent to which p300 411

interacts with FOXO3a. To do this we used a co-immunoprecipitation assay kit in which we 412

immunoprecipitated p300 from protein extracts of soleus muscles injected with an empty vector 413

or WT p300 plasmid. Following p300 protein precipitation we subsequently immunoblotted for 414

endogenous FOXO3a or FOXO1. As shown in Figure 4O and 4P, p300 interacts with both 415

19

FOXO3a and FOXO1, and this interaction is increased when p300 is overexpressed. The 416

interaction of p300 was found to be greater with FOXO1 than FOXO3a, which may therefore 417

explain our finding that p300 potentiates FOXO1 nuclear localization during nutrient 418

deprivation. 419

420

p300/CBP repression of FOXO is mediated via Akt 421

The ability of HATs to regulate FOXO signaling has previously been shown in other cell types to 422

occur through enhancing Akt-mediated repression of FOXO. This led us to question whether 423

p300/CBP could repress FOXO activation when Akt signaling is specifically inhibited. 424

Treatment of C2C12 myotubes with 10 μM LY294002 for 6 hours to inhibit PI3K/Akt signaling 425

resulted in a 40% increase in FOXO reporter activation (Figure 5A). Neither CBP nor p300 426

were able to repress FOXO reporter activation induced by LY294002 treatment. These findings 427

suggest p300/CBP may require Akt to inhibit FOXO signaling. To further test this we transfected 428

C2C12 cells with the FOXO reporter plasmid plus an empty vector, d.n.Akt, or d.n.Akt plus WT 429

p300 to determine whether p300 can repress FOXO activation when Akt activity is directly and 430

chronically inhibited. Following 3 days of differentiation luciferase activity was measured. As 431

shown in Figure 5B, transfection of d.n.Akt was sufficient to significantly increase FOXO 432

reporter activity by ~50%, which trended toward a further increase (p=0.087) in cells co-433

transfected with WT p300. This finding confirms that p300 cannot repress FOXO activation 434

when Akt activity is chronically inhibited, and suggests that p300 HAT activity may inhibit 435

FOXO through promoting FOXO inhibition by Akt. Based on this data, we hypothesized that 436

d.n.p300-induced activation of FOXO during normal conditions, in vivo (as shown in Figure 1A) 437

may have occurred through decreasing FOXO sensitivity to Akt. To test this we determined 438

20

whether increased Akt activity could overcome FOXO activation induced by d.n.p300. To do 439

this, we electroporated rat soleus muscles with the FOXO reporter plasmid and either empty 440

vector, d.n.p300, c.a.Akt, or d.n.p300 plus c.a.Akt plasmids. As shown in Figure 5C, the 441

increase in FOXO activity induced by d.n.p300 was repressed in muscles co-transfected with 442

c.a.Akt. Collectively these data demonstrate that p300 HAT activity is necessary for the normal 443

repression of FOXO in skeletal muscle under baseline conditions and that increasing HAT 444

activity is sufficient to repress FOXO activation during atrophic conditions, both of which appear 445

to be mediated via Akt-signaling. To identify whether p300 can increase FOXO phosphorylation 446

at the known Akt sites, we measured endogenous FOXO1 and FOXO3a phosphorylation from 447

soleus muscles injected with an empty vector or WT p300 plasmid. Overexpression of p300 448

increased both FOXO1 and FOXO3a phosphorylation (Figure 5D), providing further evidence 449

that p300 may increase FOXO sensitivity to Akt. Importantly, since we found in previous 450

experiments that p300 increases FOXO3a acetylation (Figure 3C), we similarly determined 451

whether p300 also acetylates FOXO1. To do this we precipitated total acetylated proteins from 452

muscles injected with either an empty vector or WT p300 plasmid, as described previously, and 453

subsequently immunoblotted for FOXO1. Similar to the effect on FOXO3a, overexpression of 454

WT p300 increased FOXO1 acetylation (Figure 5E). 455

456

457

458

459

460

461

21

DISCUSSION 462

The current study provides the first evidence to support the acetyltransferase (HAT) activities of 463

p300/CBP in regulating FOXO signaling in skeletal muscle. We demonstrate that (1) p300 HAT 464

activity is necessary to repress FOXO transcriptional activity, in vivo, during normal 465

physiological conditions, (2) increasing CBP or p300 HAT activity is sufficient to block FOXO 466

activation, in vivo, in response to skeletal muscle disuse and in C2C12 cells in response to 467

nutrient deprivation or dexamethasone treatment, (3) increased p300/CBP HAT activity can 468

repress the activation of a subset of FOXO target genes in response to catabolic stimuli, and (4) 469

p300 interacts with and acetylates both FOXO1 and FOXO3a, and differentially regulates their 470

cellular localization and transcriptional activity. Together, these findings are the first to identify 471

p300/CBP-mediated acetylation as a mechanism to regulate the FOXO transcription factors in 472

skeletal muscle. 473

The findings in the current study suggest that p300/CBP HAT activity plays an important 474

role in repressing FOXO activity in skeletal muscle during normal physiological conditions. 475

Importantly, since neither p300 nor CBP overexpression reduced basal levels of FOXO activity 476

during normal conditions, this suggests that endogenous HAT activity already maintains a 477

maximal inhibitory effect on total FOXO signaling. In contrast, because increased HAT activity 478

via either p300 or CBP overexpression repressed FOXO activation in vivo, in response to muscle 479

disuse and in vitro, in response to nutrient deprivation and dexamethasone treatment, these data 480

indicate that FOXO regulation by HAT activity is altered during catabolic conditions. The 481

insufficiency of endogenous HAT proteins to repress FOXO during these conditions may 482

potentially be explained by multiple scenarios, one of which could be a decrease in HAT 483

activity. Although the regulation of p300/CBP HAT activity is multi-factorial, their activity is 484

22

regulated in part via direct Akt-mediated phosphorylation (20), and active Akt is reduced during 485

muscle disuse, (6), nutrient deprivation (Ref) and Dex treatment (ref). In addition, acetyl-coA is 486

a substrate for p300/CBP-mediated acetylation of target substrates, and is a key intermediate in 487

numerous metabolic processes (25). Therefore, decreases in the availability of acetyl-coA during 488

catabolic conditions could also reduce p300-mediated acetylation of target proteins. 489

Alternatively, the insufficiency of HAT proteins to repress FOXO during catabolic 490

conditions could be due to an increase in histone deacetylase (HDAC) activity. HDAC proteins 491

counteract the activities of HATs by removing acetyl groups from target proteins. There are five 492

different classes of HDACs (Class I, Class IIa, Class IIb, Class III (Sirtuins or SIRTs) and Class 493

IV) which each contain multiple family members (17). Similarly, there are multiple proteins 494

which possess HAT activity, including p300, CBP, PCAF and GCN5. Since HAT and HDAC 495

proteins acetylate/deacetylate specific protein substrates, the inability of endogenous HAT 496

proteins to suppress FOXO during catabolic conditions may result from the altered expression, 497

cellular localization, and/or activity of any one or combination of the HAT or HDAC proteins. 498

While measurement of these variables was beyond the scope of the current study, other studies 499

have shown that SIRT1 (5), HDAC2 (55), HDAC4 (1, 8, 50), HDAC5 (8) and HDAC6 (50) are 500

increased in response to skeletal muscle disuse. Although we are unaware of any published data 501

to support FOXO regulation by any of these HDACs in skeletal muscle, there is certainly 502

evidence in other cell types to support FOXO regulation by the NAD+-dependent SIRTs (3, 10, 503

49). In response to low nutrient conditions when NAD+ levels are elevated, FOXO 504

deacetylation by the Sirtuins increases FOXO-dependent transcription of various genes involved 505

in both glucose metabolism (13, 29) as well as autophagy (18). Since the activities of both 506

SIRTs and HATs are regulated via bioenergetic factors (NAD+ and acetyl-coA, respectively), it 507

23

seems plausible that alterations in the energy state of the muscle during catabolic conditions 508

could dictate which FOXO-target genes are activated, through altering HAT/HDAC-mediated 509

regulation of the FOXO factors. 510

In the current study we measured the effects of p300 HAT activity on various FOXO-511

target genes known to be elevated during muscle disuse. Interestingly, increased p300 HAT 512

activity was found to repress the disuse-induced activation of some FOXO-targets (atrogin-1, 513

MuRF1 and p21), yet contributed to the increased expression of others (Gadd45α, 4E-BP1 and 514

cathepsin-L). Subsequent experiments further found that p300 also differentially regulated the 515

activity and localization of the FOXO homologues, which could potentially explain the 516

differential effect of p300 and its HAT activity on FOXO target gene expression. Although the 517

relative contribution of each endogenous FOXO homologue to the regulation of these genes in 518

skeletal muscle during physiological muscle wasting is not known, FOXO overexpression and 519

transgenic studies have yielded important information in this regard. Several studies have now 520

demonstrated that FOXO3a is sufficient to increase atrogin-1, MuRF1 and LC3 gene 521

transcription (43, 54), both in C2C12s and in whole muscle. In contrast, the data is conflicting 522

on whether FOXO1 is sufficient to increase atrogin-1 and/or MuRF1 in skeletal muscle (23, 48, 523

54). Importantly, however, cathepsin L and Gadd45α mRNA levels are increased in the skeletal 524

muscle of FOXO1 transgenic mice and decreased in mice transgenically expressing d.n.FOXO1 525

(56). These genetic studies therefore suggest that atrogin-1 and MuRF1 are primary targets of 526

FOXO3a and that cathepsin L and Gadd45α are primary targets of FOXO1. Given our collective 527

findings that (a) p300 HAT activity represses FOXO3a transcriptional activity and nuclear 528

localization and represses atrogin-1 and MuRF1 gene expression and (b) p300 increases FOXO1 529

nuclear localization and contributes to the increased gene expression of cathepsin-L and 530

24

Gadd45α, it may be speculated that p300 regulates these genes through differentially regulating 531

FOXO3a- and FOXO1-dependent gene transcription. 532

It is important to mention however, that p300 also regulates histone configuration, can act 533

as a transcriptional co-activator, and may regulate additional transcription factors other than 534

FOXO. Therefore the changes in mRNA levels observed with the p300 constructs may also 535

reflect changes in these variables. However, our use of the atrogin-1 promoter reporter plasmid 536

circumnavigates at least one of these issues. Since plasmid DNA constructs remain 537

extrachromosomal, their regulation does not therefore depend upon an open histone 538

configuration for active gene transcription (as does genomic DNA). Therefore, at least for 539

atrogin-1, p300-mediated regulation of its promoter activity is not mediated through changes in 540

histone configuration. 541

While the current study provides evidence that p300/CBP-mediated repression of FOXO 542

may require intact Akt signaling, p300/CBP overexpression still repressed the physiological 543

activation of FOXO during skeletal muscle disuse. Therefore, although the levels of active Akt 544

are reduced, in vivo, during periods of muscle disuse (6), these reduced levels of Akt were 545

presumably sufficient for overexpressed WT p300/CBP to repress the disuse-induced activation 546

of FOXO in the current study. Furthermore, since reduction of endogenous HAT activity via 547

expression of d.n.p300 in control muscle was sufficient to activate FOXO, this also demonstrates 548

that FOXO can be activated without directly manipulating Akt levels. Collectively, these 549

findings suggest that it may be possible to therapeutically manipulate Akt-mediated repression of 550

FOXO indirectly, via targeting HAT activity, which would have important ramifications for the 551

muscle wasting field. 552

25

In summary these findings demonstrate that p300/CBP acetyltransferase activity is both 553

necessary and sufficient to repress FOXO transcriptional activity in skeletal muscle, in vivo. 554

Furthermore, this study offers new insight into the differential regulation of the FOXO 555

homologues in skeletal muscle and highlights new therapeutic possibilities for blocking specific 556

FOXO target-genes during conditions of muscle wasting. 557

558

GRANTS 559

This work was supported by National Institute of Arthritis and Musculoskeletal and Skin 560

Diseases Grant R03AR056418 (to A. R. Judge). S. M. Senf is supported by a T32 from the 561

National Institute of Child Health and Human Development Grant T32-HD-043730. 562

563

DISCLOSURES 564

No conflicts of interest are declared by the author(s). 565

566 567 568 569 570 571 572 573 574 575 576 577 578 579 580 581 582

26

References 583 584 1. Bodine SC, Latres E, Baumhueter S, Lai VK, Nunez L, Clarke BA, Poueymirou 585 WT, Panaro FJ, Na E, Dharmarajan K, Pan ZQ, Valenzuela DM, DeChiara TM, Stitt TN, 586 Yancopoulos GD, and Glass DJ. Identification of ubiquitin ligases required for skeletal muscle 587 atrophy. Science 294: 1704-1708, 2001. 588 2. Brunet A, Bonni A, Zigmond MJ, Lin MZ, Juo P, Hu LS, Anderson MJ, Arden KC, 589 Blenis J, and Greenberg ME. Akt promotes cell survival by phosphorylating and inhibiting a 590 Forkhead transcription factor. Cell 96: 857-868, 1999. 591 3. Brunet A, Sweeney LB, Sturgill JF, Chua KF, Greer PL, Lin Y, Tran H, Ross SE, 592 Mostoslavsky R, Cohen HY, Hu LS, Cheng HL, Jedrychowski MP, Gygi SP, Sinclair DA, 593 Alt FW, and Greenberg ME. Stress-dependent regulation of FOXO transcription factors by the 594 SIRT1 deacetylase. Science 303: 2011-2015, 2004. 595 4. Caiozzo VJ, Wu YZ, Baker MJ, and Crumley R. Effects of Denervation on Cell Cycle 596 Control in Laryngeal Muscle. Arch Otolaryngol Head Neck Surg 130: 1056-1068, 2004. 597 5. Chabi B, Adhihetty PJ, O'Leary MF, Menzies KJ, and Hood DA. Relationship 598 between Sirt1 expression and mitochondrial proteins during conditions of chronic muscle use 599 and disuse. J Appl Physiol 107: 1730-1735, 2009. 600 6. Childs TE, Spangenburg EE, Vyas DR, and Booth FW. Temporal alterations in 601 protein signaling cascades during recovery from muscle atrophy. Am J Physiol Cell Physiol 285: 602 C391-398, 2003. 603 7. Clavel S, Siffroi-Fernandez S, Coldefy AS, Boulukos K, Pisani DF, and Derijard B. 604 Regulation of the intracellular localization of Foxo3a by stress-activated protein kinase signaling 605 pathways in skeletal muscle cells. Molecular and cellular biology 30: 470-480. 606 8. Cohen TJ, Waddell DS, Barrientos T, Lu Z, Feng G, Cox GA, Bodine SC, and Yao 607 TP. The histone deacetylase HDAC4 connects neural activity to muscle transcriptional 608 reprogramming. The Journal of biological chemistry 282: 33752-33759, 2007. 609 9. Crossland H, Constantin-Teodosiu D, Gardiner SM, Constantin D, and Greenhaff 610 PL. A potential role for Akt/FOXO signalling in both protein loss and the impairment of muscle 611 carbohydrate oxidation during sepsis in rodent skeletal muscle. J Physiol 586: 5589-5600, 2008. 612 10. Daitoku H, Hatta M, Matsuzaki H, Aratani S, Ohshima T, Miyagishi M, Nakajima 613 T, and Fukamizu A. Silent information regulator 2 potentiates Foxo1-mediated transcription 614 through its deacetylase activity. Proceedings of the National Academy of Sciences of the United 615 States of America 101: 10042-10047, 2004. 616 11. Dansen TB, Smits LM, van Triest MH, de Keizer PL, van Leenen D, Koerkamp 617 MG, Szypowska A, Meppelink A, Brenkman AB, Yodoi J, Holstege FC, and Burgering 618 BM. Redox-sensitive cysteines bridge p300/CBP-mediated acetylation and FoxO4 activity. 619 Nature chemical biology 5: 664-672, 2009. 620 12. Eberharter A and Becker PB. Histone acetylation: a switch between repressive and 621 permissive chromatin. Second in review series on chromatin dynamics. EMBO Rep 3: 224-229, 622 2002. 623 13. Frescas D, Valenti L, and Accili D. Nuclear trapping of the forkhead transcription factor 624 FoxO1 via Sirt-dependent deacetylation promotes expression of glucogenetic genes. The Journal 625 of biological chemistry 280: 20589-20595, 2005. 626 14. Fukuoka M, Daitoku H, Hatta M, Matsuzaki H, Umemura S, and Fukamizu A. 627 Negative regulation of forkhead transcription factor AFX (Foxo4) by CBP-induced acetylation. 628 International journal of molecular medicine 12: 503-508, 2003. 629

27

15. Furuyama T, Kitayama K, Yamashita H, and Mori N. Forkhead transcription factor 630 FOXO1 (FKHR)-dependent induction of PDK4 gene expression in skeletal muscle during 631 energy deprivation. The Biochemical journal 375: 365-371, 2003. 632 16. Giresi PG, Stevenson EJ, Theilhaber J, Koncarevic A, Parkington J, Fielding RA, 633 and Kandarian SC. Identification of a molecular signature of sarcopenia. Physiological 634 genomics 21: 253-263, 2005. 635 17. Haberland M, Montgomery RL, and Olson EN. The many roles of histone 636 deacetylases in development and physiology: implications for disease and therapy. Nature 637 reviews 10: 32-42, 2009. 638 18. Hariharan N, Maejima Y, Nakae J, Paik J, Depinho RA, and Sadoshima J. 639 Deacetylation of FoxO by Sirt1 Plays an Essential Role in Mediating Starvation-Induced 640 Autophagy in Cardiac Myocytes. Circulation research 107: 1470-1482. 641 19. Hasselgren PO. Ubiquitination, phosphorylation, and acetylation--triple threat in muscle 642 wasting. J Cell Physiol 213: 679-689, 2007. 643 20. Huang WC and Chen CC. Akt phosphorylation of p300 at Ser-1834 is essential for its 644 histone acetyltransferase and transcriptional activity. Molecular and cellular biology 25: 6592-645 6602, 2005. 646 21. Ito A, Lai CH, Zhao X, Saito S, Hamilton MH, Appella E, and Yao TP. p300/CBP-647 mediated p53 acetylation is commonly induced by p53-activating agents and inhibited by 648 MDM2. EMBO J 20: 1331-1340, 2001. 649 22. Judge AR, Koncarevic A, Hunter RB, Liou HC, Jackman RW, and Kandarian SC. 650 Role for IkappaBalpha, but not c-Rel, in skeletal muscle atrophy. American journal of physiology 651 292: C372-382, 2007. 652 23. Kamei Y, Miura S, Suzuki M, Kai Y, Mizukami J, Taniguchi T, Mochida K, Hata T, 653 Matsuda J, Aburatani H, Nishino I, and Ezaki O. Skeletal muscle FOXO1 (FKHR) transgenic 654 mice have less skeletal muscle mass, down-regulated Type I (slow twitch/red muscle) fiber 655 genes, and impaired glycemic control. The Journal of biological chemistry 279: 41114-41123, 656 2004. 657 24. Kamei Y, Mizukami J, Miura S, Suzuki M, Takahashi N, Kawada T, Taniguchi T, 658 and Ezaki O. A forkhead transcription factor FKHR up-regulates lipoprotein lipase expression 659 in skeletal muscle. FEBS letters 536: 232-236, 2003. 660 25. Kim SC, Sprung R, Chen Y, Xu Y, Ball H, Pei J, Cheng T, Kho Y, Xiao H, Xiao L, 661 Grishin NV, White M, Yang XJ, and Zhao Y. Substrate and functional diversity of lysine 662 acetylation revealed by a proteomics survey. Molecular cell 23: 607-618, 2006. 663 26. Latres E, Amini AR, Amini AA, Griffiths J, Martin FJ, Wei Y, Lin HC, 664 Yancopoulos GD, and Glass DJ. Insulin-like growth factor-1 (IGF-1) inversely regulates 665 atrophy-induced genes via the phosphatidylinositol 3-kinase/Akt/mammalian target of rapamycin 666 (PI3K/Akt/mTOR) pathway. The Journal of biological chemistry 280: 2737-2744, 2005. 667 27. Lecker SH, Jagoe RT, Gilbert A, Gomes M, Baracos V, Bailey J, Price SR, Mitch 668 WE, and Goldberg AL. Multiple types of skeletal muscle atrophy involve a common program 669 of changes in gene expression. Faseb J 18: 39-51, 2004. 670 28. Lee SW, Dai G, Hu Z, Wang X, Du J, and Mitch WE. Regulation of muscle protein 671 degradation: coordinated control of apoptotic and ubiquitin-proteasome systems by 672 phosphatidylinositol 3 kinase. J Am Soc Nephrol 15: 1537-1545, 2004. 673

28

29. Liu Y, Dentin R, Chen D, Hedrick S, Ravnskjaer K, Schenk S, Milne J, Meyers DJ, 674 Cole P, Yates J, 3rd, Olefsky J, Guarente L, and Montminy M. A fasting inducible switch 675 modulates gluconeogenesis via activator/coactivator exchange. Nature 456: 269-273, 2008. 676 30. Machida S and Booth FW. Changes in signalling molecule levels in 10-day hindlimb 677 immobilized rat muscles. Acta Physiol Scand 183: 171-179, 2005. 678 31. Marmorstein R. Structure and function of histone acetyltransferases. Cell Mol Life Sci 679 58: 693-703, 2001. 680 32. Matsuzaki H, Daitoku H, Hatta M, Aoyama H, Yoshimochi K, and Fukamizu A. 681 Acetylation of Foxo1 alters its DNA-binding ability and sensitivity to phosphorylation. 682 Proceedings of the National Academy of Sciences of the United States of America 102: 11278-683 11283, 2005. 684 33. McClung JM, Judge AR, Powers SK, and Yan Z. p38 MAPK links oxidative stress to 685 autophagy-related gene expression in cachectic muscle wasting. American journal of physiology 686 298: C542-549. 687 34. Motta MC, Divecha N, Lemieux M, Kamel C, Chen D, Gu W, Bultsma Y, 688 McBurney M, and Guarente L. Mammalian SIRT1 represses forkhead transcription factors. 689 Cell 116: 551-563, 2004. 690 35. Perrot V and Rechler MM. The coactivator p300 directly acetylates the forkhead 691 transcription factor Foxo1 and stimulates Foxo1-induced transcription. Molecular endocrinology 692 (Baltimore, Md 19: 2283-2298, 2005. 693 36. Qiang L, Banks AS, and Accili D. Uncoupling of acetylation from phosphorylation 694 regulates FoxO1 function independent of its subcellular localization. The Journal of biological 695 chemistry 285: 27396-27401. 696 37. Ramaswamy S, Nakamura N, Sansal I, Bergeron L, and Sellers WR. A novel 697 mechanism of gene regulation and tumor suppression by the transcription factor FKHR. Cancer 698 Cell 2: 81-91, 2002. 699 38. Rana ZA, Ekmark M, and Gundersen K. Coexpression after electroporation of 700 plasmid mixtures into muscle in vivo. Acta Physiol Scand 181: 233-238, 2004. 701 39. Reed SA, Senf SM, Cornwell EW, Kandarian SC, and Judge AR. Inhibition of 702 IkappaB kinase alpha (IKKalpha) or IKKbeta (IKKbeta) plus forkhead box O (Foxo) abolishes 703 skeletal muscle atrophy. Biochem Biophys Res Commun. 704 40. Sacheck JM, Hyatt JP, Raffaello A, Jagoe RT, Roy RR, Edgerton VR, Lecker SH, 705 and Goldberg AL. Rapid disuse and denervation atrophy involve transcriptional changes similar 706 to those of muscle wasting during systemic diseases. FASEB J 21: 140-155, 2007. 707 41. Sadoul K, Boyault C, Pabion M, and Khochbin S. Regulation of protein turnover by 708 acetyltransferases and deacetylases. Biochimie 90: 306-312, 2008. 709 42. Salih DA and Brunet A. FoxO transcription factors in the maintenance of cellular 710 homeostasis during aging. Curr Opin Cell Biol 20: 126-136, 2008. 711 43. Sandri M, Sandri C, Gilbert A, Skurk C, Calabria E, Picard A, Walsh K, Schiaffino 712 S, Lecker SH, and Goldberg AL. Foxo transcription factors induce the atrophy-related 713 ubiquitin ligase atrogin-1 and cause skeletal muscle atrophy. Cell 117: 399-412, 2004. 714 44. Schulze PC, Fang J, Kassik KA, Gannon J, Cupesi M, MacGillivray C, Lee RT, and 715 Rosenthal N. Transgenic overexpression of locally acting insulin-like growth factor-1 inhibits 716 ubiquitin-mediated muscle atrophy in chronic left-ventricular dysfunction. Circulation research 717 97: 418-426, 2005. 718

29

45. Senf SM, Dodd SL, and Judge AR. FOXO Signaling is Required for Disuse Muscle 719 Atrophy and is Directly Regulated by Hsp70. American journal of physiology, 2009. 720 46. Senf SM, Dodd SL, McClung JM, and Judge AR. Hsp70 overexpression inhibits NF-721 kappaB and Foxo3a transcriptional activities and prevents skeletal muscle atrophy. Faseb J 22: 722 3836-3845, 2008. 723 47. Spange S, Wagner T, Heinzel T, and Kramer OH. Acetylation of non-histone proteins 724 modulates cellular signalling at multiple levels. Int J Biochem Cell Biol 41: 185-198, 2009. 725 48. Stitt TN, Drujan D, Clarke BA, Panaro F, Timofeyva Y, Kline WO, Gonzalez M, 726 Yancopoulos GD, and Glass DJ. The IGF-1/PI3K/Akt pathway prevents expression of muscle 727 atrophy-induced ubiquitin ligases by inhibiting FOXO transcription factors. Molecular cell 14: 728 395-403, 2004. 729 49. Sundaresan NR, Gupta M, Kim G, Rajamohan SB, Isbatan A, and Gupta MP. Sirt3 730 blocks the cardiac hypertrophic response by augmenting Foxo3a-dependent antioxidant defense 731 mechanisms in mice. J Clin Invest 119: 2758-2771, 2009. 732 50. Tang H, Macpherson P, Marvin M, Meadows E, Klein WH, Yang XJ, and Goldman 733 D. A histone deacetylase 4/myogenin positive feedback loop coordinates denervation-dependent 734 gene induction and suppression. Mol Biol Cell 20: 1120-1131, 2009. 735 51. Van Der Heide LP, Hoekman MF, and Smidt MP. The ins and outs of FoxO shuttling: 736 mechanisms of FoxO translocation and transcriptional regulation. The Biochemical journal 380: 737 297-309, 2004. 738 52. van der Heide LP and Smidt MP. Regulation of FoxO activity by CBP/p300-mediated 739 acetylation. Trends Biochem Sci 30: 81-86, 2005. 740 53. van der Horst A, Tertoolen LG, de Vries-Smits LM, Frye RA, Medema RH, and 741 Burgering BM. FOXO4 is acetylated upon peroxide stress and deacetylated by the longevity 742 protein hSir2(SIRT1). The Journal of biological chemistry 279: 28873-28879, 2004. 743 54. Waddell DS, Baehr LM, van den Brandt J, Johnsen SA, Reichardt HM, Furlow JD, 744 and Bodine SC. The glucocorticoid receptor and FOXO1 synergistically activate the skeletal 745 muscle atrophy-associated MuRF1 gene. Am J Physiol Endocrinol Metab 295: E785-797, 2008. 746 55. Wu CL, Kandarian SC, and Jackman RW. Identification of Genes that Elicit Disuse 747 Muscle Atrophy via the Transcription Factors p50 and Bcl-3. PLoS One 6: e16171. 748 56. Yamazaki Y, Kamei Y, Sugita S, Akaike F, Kanai S, Miura S, Hirata Y, Troen BR, 749 Kitamura T, Nishino I, Suganami T, Ezaki O, and Ogawa Y. The cathepsin L gene is a direct 750 target of FOXO1 in skeletal muscle. Biochemical Journal 427: 171-178, 2010. 751 57. Zhao J, Brault JJ, Schild A, Cao P, Sandri M, Schiaffino S, Lecker SH, and 752 Goldberg AL. FoxO3 coordinately activates protein degradation by the autophagic/lysosomal 753 and proteasomal pathways in atrophying muscle cells. Cell Metab 6: 472-483, 2007. 754 58. Zhou BP, Hu MC, Miller SA, Yu Z, Xia W, Lin SY, and Hung MC. HER-2/neu 755 blocks tumor necrosis factor-induced apoptosis via the Akt/NF-kappaB pathway. J Biol Chem 756 275: 8027-8031, 2000. 757 758 759 760 761 762 763

764

30

FIGURE LEGENDS 765

Figure 1- p300 acetyltransferase activity is necessary and sufficient to repress FOXO 766

transcriptional activity in skeletal muscle. A and B: Foxo-dependent luciferase reporter 767

activity from weight bearing and 3-day cast immobilized solei injected with WT p300 or 768

d.n.p300 (lacks HAT activity) (A) or WT CBP (B), or the respective control plasmids (empty 769

vector; EV). A representative western blot for p300 from EV, WT p300 and d.n.p300 injected 770

muscles is shown in A. Data are reported as mean ± SEM, normalized to the weight bearing, 771

empty vector injected group, and N=at least 5 muscles/group C: C2C12 myoblasts were 772

transfected with a FOXO-responsive reporter, pRL-TK-Renilla, and either an empty vector, WT 773

p300 or WT CBP expression plasmid. Following 6 days of differentiation myotubes were 774

treated with vehicle or 1µM dexamethasone (DEX) for 6 hours and assayed for luciferase 775

activity. Data represent N=3 and are reported as mean ± SEM, normalized to the absolute 776

control group. Experiments were repeated at least three times. Significance was established at 777

p<0.05. *Significantly different from absolute control group. †Significantly different from empty 778

vector within respective treatment group. 779

780

Figure 2- p300 HAT activity differentially regulates the transcription of FOXO-target 781

genes during muscle disuse. A: Relative atrogin-1 mRNA levels from weight bearing or 3-day 782

cast immobilized solei injected with an empty vector, WT p300 or d.n.p300 plasmid. B: 783

Atrogin-1 promoter reporter activity from weight bearing or 3-day cast immobilized rat solei 784

injected with an atrogin-1 promoter luciferase reporter plus an Empty Vector, WT p300 or 785

d.n.p300 plasmid. C: Atrogin-1 promoter reporter activity from weight bearing rat solei co-786

injected with an empty vector or d.n.p300 plus either an empty vector or a d.n.FOXO construct. 787

31

D & E: Relative mRNA levels of (D) MuRF1, p21, 4E-BP1, Gadd45α, cathepsin-L, LC3b, 18S 788

and, (E) Foxo1, Foxo3a, and Foxo4 from rat solei injected with an empty vector, WT p300 or 789

d.n.p300 and exposed to weight bearing or 3-days of cast immobilization. All data are reported 790

as mean ± SEM, normalized to the absolute control group, N=at least 5 muscles/group. 791

Significance was established at p<0.05. *Significantly different from absolute control group. 792

†Significantly different from empty vector within respective treatment group. 793

794

Figure 3- p300 HAT activity differentially regulates the transcriptional activity of the 795

FOXO homologues. A-E: Rat soleus muscles were injected and electroporated with a FOXO-796

responsive luciferase reporter plus an empty vector (EV), FOXO1, FOXO3a or FOXO4 797

expression plasmid, each with an EV, WT p300, or d.n.p300 plasmid. A: Representative western 798

blots for total FOXO1, FOXO3a and FOXO4 overexpression. B: Seven days following plasmid 799

injection muscles were removed and assayed for FOXO-dependent luciferase activity. All data 800

are reported as mean ± SEM from at least 6 muscles/group, normalized to the absolute control-801

injected group and significance was established at p<0.05. *Significantly different from absolute 802

control group (EV only). †Significantly different from FOXO + EV within respective FOXO 803

group. #Significantly different. C: Endogenous FOXO3a acetylation was determined in soleus 804

muscles injected with an empty vector (EV), WT p300 or d.n.p300 plasmid via 805

immunoprecipitation (IP) of total acetylated proteins from protein extracts using an anti-acetyl-806

lysine antibody followed by western blot for FOXO3a. Experiments were independently 807

repeated three times. D: Representative western blot showing FOXO3a protein levels in soleus 808

muscles injected with FOXO3a plus an EV, WT p300 or d.n.p300, using α-tubulin as loading 809

control. E: FOXO activity (underlined in Figure 3B) normalized to total FOXO3a protein levels 810

32

from muscles injected with FOXO3a plus either an empty vector, WT p300 or d.n.p300 (from 811

Figure 3D). 812

813

Figure 4- p300 and CBP differentially regulate FOXO3a and FOXO1 cellular localization. 814

A-L: FOXO1 and FOXO3a localization in C2C12 cells transfected with FOXO3a-DsRed or 815

FOXO1-GFP plus an empty vector (EV), WT p300 or WT CBP plasmid, differentiated for 4 816

days and either left in differentiation media (control) or nutrient deprived by replacing media 817

with HBSS for 2 hours. FOXO3a-DsRed localization was visualized using a Rhodamine (Red) 818

filter and was merged with Dapi-stained (blue) nuclei (A-F). Arrows in B point to FOXO3a-819

DsRed positive nuclei, which are pink in the merged image. FOXO1-GFP positive myotubes 820

were visualized using a GFP (green) filter and were merged with Dapi-stained (blue) nuclei (G-821

L). Arrows in H and J point to nuclei that are FOXO1-GFP positive, and which are light blue in 822

merged images. M: C2C12 myoblasts transfected with a FOXO-responsive reporter, pRL-TK-823

Renilla, and an EV, WT p300 or WT CBP expression plasmid. Following 4 days of 824

differentiation myotubes were left in differentiation media (control) or nutrient deprived (HBSS) 825

for 6 hours and harvested for luciferase activity. Data are expressed as mean ± SEM, and are 826

normalized to the absolute control group. N: Atrogin-1 gene expression following 6 hours of 827

nutrient deprivation (HBSS) of C2C12 cells. Myoblasts were transfected with either an EV or 828

WT CBP and differentiated for 4-days prior to treatment. Data are expressed as mean ± SEM, 829

and are normalized to the respective control group. All data represent N=3, and experiments 830

were independently repeated at least three times. Significance was established at p<0.05. 831

*Significantly different from the respective control group. †Significantly different from EV + 832

HBSS group. O and P: The ability of p300 to interact with endogenous FOXO3a (O) and 833

33

FOXO1 (P) was determined in soleus muscles injected with either an empty vector or WT p300 834

expression plasmid. Equal amounts of protein extract were used to immunoprecipitate (IP) p300, 835

followed by subsequent immunoblot for either FOXO3a or FOXO1. Western blots for 836

endogenous FOXO3a and FOXO1 indicate the relative input for IP experiments. Experiments 837

were independently repeated three times. 838

839

Figure 5- HAT-induced repression of FOXO is mediated via Akt. 840

A: C2C12 cells were transfected with a FOXO-responsive reporter, pRL-TK-Renilla, and either 841

an empty vector (EV), WT p300 or WT CBP expression plasmid. Following 4 days of 842

differentiation myotubes were treated with either vehicle (ethanol) or the PI3Kinase inhibitor 843

LY294002 (10uM) for 6 hours, and harvested for luciferase activity. Data are normalized to their 844

respective vehicle treated group, and are therefore expressed as (+/-) LY294002. B: C2C12 cells 845

were transfected with a FOXO-responsive reporter, pRL-TK-Renilla, and either an EV, WT 846

p300, d.n.Akt, or WT p300 + d.n.Akt. Following 3 days of differentiation, myotubes were 847

harvested for luciferase activity. Data are normalized to the absolute control group. C: Rat 848

soleus muscles were injected and electroporated with a FOXO-responsive reporter plus either an 849

EV, d.n.p300, c.a.Akt, or c.a.Akt + d.n.p300. Seven days following injections, muscles were 850

removed and harvested for luciferase activity. Data are normalized to the respective empty 851

vector group (black bars) within the EV or c.a.Akt groups. All cell culture data represent N=3 852

and are reported as mean ± SEM, and were repeated at least three times. Significance was 853

established at p<0.05. *Significantly different from absolute control group. D: Representative 854

western blots showing phospho- and total FOXO1 and FOXO3a from soleus muscles injected 855

with either an empty vector or WT p300 plasmid. E: Endogenous FOXO1 acetylation from 856

34

muscle extracts in (D) was determined via immunoprecipitation (IP) of total acetylated proteins 857

using an anti-acetyl-lysine antibody followed by western blot for FOXO1. Experiments 858

represent N=3. 859

860

Figure 6- Proposed regulation of FOXO by p300/CBP acetyltransferase (HAT) activity. 861

A: During normal physiological conditions, HAT proteins acetylate FOXO and promote FOXO 862

retention in the cytosol by Akt. B: In response to catabolic conditions, disruptions in both Akt 863

and HAT signaling contribute to FOXO nuclear localization and transcriptional activation of 864

target-genes. 865

866

0.0

2.0

4.0

6.0

8.0

10.0

12.0

14.0

16.0 Empty VectorWT p300d.n.p300

WB Imm

*

* †

*

FOXO

(6xD

BE)

Rep

orte

r(R

elat

ive

luci

fera

se a

ctiv

ity)

A B

250kDa

WB:p300

Figure 1

EV WTp300 d.n.p300

Vehicle Dex0.0

0.5

1.0

1.5

2.0

2.5

*

Empty VectorWT p300

WT CBP

FOXO

(6xD

BE)

Rep

orte

r(R

elat

ive

luci

fera

se a

ctiv

ity)

C

WB Imm0.0

0.5

1.0

1.5

2.0

2.5

Empty VectorWT CBP

*

FOXO

(6xD

BE)

Rep

orte

r(R

elat

ive

luci

fera

se a

ctiv

ity)

WB Imm WB Imm WB Imm WB Imm WB Imm WB Imm WB Imm0.0

0.5

1.0

1.5

2.0

2.5

3.0

3.5

4.0

4.5

MuRF1 Cathepsin-L4E-BP1 18Sp21

*

LC3Gadd45�

*

*†

* **

*

†*

*†

*

*†

* * Empty VectorWT p300d.n.p300

**

*

Rel

ativ

e m

RN

A le

vel

0.0

1.0

2.0

3.0

4.0

5.0

6.0

Empty VectorWT p300d.n.p300

WB Imm

*

*

Atro

gin-

1 m

RN

A le

vel

0.0

1.0

2.0

3.0

4.0

5.0

Empty VectorWT p300d.n.p300

WB Imm

*

*

*

*

Atro

gin-

1 Pr

omot

er R

epor

ter

(Rel

ativ

e lu

cife

rase

act

ivity

)

0.0

1.0

2.0

3.0

4.0

5.0Empty Vectord.n.p300

EV d.n.FOXO

*

Atro

gin-

1 Pr

omot

er R

epor

ter

(Rel

ativ

e lu

cife

rase

act

ivity

)

Figure 2

CA B

D

WB Imm WB Imm WB Imm0.0

0.5

1.0

1.5

2.0

2.5Empty VectorWT p300d.n.p300

* **

Foxo1 Foxo3a Foxo4

* **

*

*

*

Rel

ativ

e m

RN

A le

vel

E

A

FOXO1

FOXO3a

- +

- +

FOXO4 - +

100kDa

75kDa

WB:FOXO1

WB:FOXO3a

WB:FOXO4

75kDa

100kDa75kDa

D

EV FOXO1 FOXO3a FOXO40.02.04.06.08.0

10.012.014.016.018.020.0

d.n.p300

Empty VectorWT p300

*

* *

#

FOXO

(6XD

BE)

Rep

orte

r(R

elat

ive

luci

fera

se a

ctiv

ity)

B

50kDa

C

FOXO3a

FOXO3a

Tubulin

FOXO3aWT p300

d.n.p300

Tubulin

EV

WTp30

0

d.n.p300

0.0

1.0

2.0

3.0

4.0

5.0

FOXO

Rep

orte

r Act

ivity

/Ec

topi

c FO

XO3a

pro

tein

EEV

WTp300

d.n.p300

IP:Acetyl-K

WB:FOXO3a

Input:

WB:

FOXO3a

Figure 3

EV

EV

EV

WT p300

WT CBP

0.00

0.25

0.50

0.75

1.00

1.25

1.50

Control6 hr HBSS

**

*

FOXO

(6xD

BE)

Rep

orte

r(R

elat

ive

luci

fera

se a

ctiv

ity)

Control Nutrient Deprivation

A B

C D

DapiFOXO3a Merge Dapi Merge

DapiFOXO1 Merge DapiFOXO1 Merge

B

E

G H

I J

EV

WT CBP

0.0

0.5

1.0

1.5

2.0

2.5

3.0

Control6 hr HBSS†

*

*

Rel

ativ

e At

rogi

n-1

mR

NA

(Fol

d)

M

N

K L

F

Figure 4

IP: p300

Input

FOXO3a

-WT p300

WB:FOXO3a

WB:FOXO3a

WTCBP

WTp300

EV

WTCBP

WTp300

EV

IP: p300

WT p300

WB:FOXO1

Input WB:FOXO1

O P

+ - +

EV d.n.Akt0.0

0.5

1.0

1.5

2.0

Empty VectorWT p300

*

*

FOXO

(6xD

BE)

Rep

orte

r(R

elat

ive

luci

fera

se a

ctiv

ity)

Figure 5BA

EV c.a.Akt0.0

1.0

2.0

3.0

4.0

5.0

Empty Vectord.n.p300

*

FOXO

(6xD

BE)

Rep

orte

r(R

elat

ive

luci

fera

se a

ctiv

ity)

EV

WT p300

WT CBP

0.00

0.25

0.50

0.75

1.00

1.25

1.50

1.75

(+/-) LY294002

FOXO

(6xD

BE)

Rep

orte

r(R

elat

ive

luci

fera

se a

ctiv

ity)

C

D100 kDa

75 kDa100 kDa

75 kDa

WB:p-FOXO3a

WB:FOXO3a

100 kDa

75 kDa

WB:p-FOXO1

WB:FOXO1

WT p300 - +

WT p300 - +

100 kDa

75 kDa

100 kDa

75 kDa

50 kDa

WB:FOXO1

IgG

IP: Acetyl-KE

AktNormal�physiological�

conditions AktCatabolic�conditions

Figure 6

A B

HATs

FOXO

X

Ac

FOXOAc

P

FOXO

Atrogin�1/MAFbxMuRF1p21

FOXO

HATs

AcAc