ER stress and cancer: The FOXO forkhead transcription factor link

ER stress and cancer: The FOXO forkhead transcription factor link

Accepted Manuscript ER stress and cancer: The FOXO forkhead transcription factor link Glowi Alasiri, Lavender Yuen-Nam Fan, Stefania Zona, Isabella Ga...

1MB Sizes 1 Downloads 110 Views

Accepted Manuscript ER stress and cancer: The FOXO forkhead transcription factor link Glowi Alasiri, Lavender Yuen-Nam Fan, Stefania Zona, Isabella Galeno Goldsbrough, Hui-Ling Ke, Holger Werner Auner, Eric Wing-Fai Lam PII:

S0303-7207(17)30296-4

DOI:

10.1016/j.mce.2017.05.027

Reference:

MCE 9958

To appear in:

Molecular and Cellular Endocrinology

Received Date: 24 March 2017 Revised Date:

17 May 2017

Accepted Date: 24 May 2017

Please cite this article as: Alasiri, G., Fan, L.Y.-N., Zona, S., Goldsbrough, I.G., Ke, H.-L., Auner, H.W., Lam, E.W.-F., ER stress and cancer: The FOXO forkhead transcription factor link, Molecular and Cellular Endocrinology (2017), doi: 10.1016/j.mce.2017.05.027. This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

ACCEPTED MANUSCRIPT 1

ER stress and cancer: the FOXO forkhead transcription factor link

2 3 4

Glowi Alasiri1,3, Lavender Yuen-Nam Fan1,3, Stefania Zona1, Isabella Galeno Goldsbrough1, Hui-Ling Ke1, Holger Werner Auner2*, Eric Wing-Fai Lam1* 1

Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London W12 0NN, UK.

15

College London, Hammersmith Hospital Campus, Du Cane Road, London W12

16

0NN,

17

[email protected];

18

Holger W. Auner, Department of Medicine, Imperial College London, Hammersmith

19

Hospital Campus, Du Cane Road, London W12 0NN, UK Phone: +44-20-3313-4017;

20

E-mail: [email protected];

21

Abstract

22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43

The endoplasmic reticulum (ER) is a cellular organelle with central roles in maintaining proteostasis due to its involvement in protein synthesis, folding, quality control, distribution and degradation. The accumulation of misfolded proteins in the ER lumen causes ‘ER stress’ and threatens overall cellular proteostasis. To restore ER homeostasis, cells evoke an evolutionarily conserved adaptive signalling and gene expression network collectively called the ‘unfolded protein response (UPR)’, a complex biological process which aims to restore proteostasis. When ER stress is overwhelming and beyond rectification, the normally pro-survival UPR can shift to induce cell termination. Emerging evidence from mammalian, fly and nematode worm systems reveals that the FOXO Forkhead proteins integrate upstream ER stress and UPR signals with the transcriptional machinery to decrease translation, promote cell survival/termination and increase the levels of ER-resident chaperones and of ER-associated degradation (ERAD) components to restore ER homeostasis. The high rates of protein synthesis/translation associated with cancer cell proliferation and metabolism, as well as mutations resulting in aberrant proteins, also induce ER stress and the UPR. While the pro-survival side of the UPR underlies its ability to sustain and promote cancers, its apoptotic functions can be exploited for cancer therapies by offering the chance to ‘flick the proteostatic switch’. To this end, further studies are required to fully reevaluate the roles and regulation of these UPR signalling molecules, including FOXO proteins and their targets, in cancer initiation and progression as well as the effects on inhibiting their functions in cancer cells. This information will help to establish these UPR signalling molecules as possible therapeutic targets and putative biomarkers in cancers.

RI PT

5 6 7 8 9 10 11 12 13 14

2

Department of Medicine, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, UK. Contributed equally and are joint first authors

SC

3

+44-20-7594-2810;

TE D

Phone:

AC C

EP

UK

M AN U

*Correspondence: Eric W.-F. Lam, Department of Surgery and Cancer, Imperial

1

Fax:

+44-20-8383-5830;

E-mail:

ACCEPTED MANUSCRIPT

81

TE D

M AN U

SC

RI PT

AKT, Protein Kinase B; AKT1, Protein Kinase B Alpha; AMPK, AMP-activated protein kinase; ASK1, Apoptosis Signal Regulated Kinase 1; ATF4, Activating transcription factor 4; ATF6, Activating Transcription Factor 6α; ATG5,Autophagy protein 5; ATG7,Autophagy protein 7 ; ATG10, Autophagy protein 10; Bak, BCL2 Antagonist/Killer 1; BAX,BCL2 Associated X; BBC3, p53-upregulated modulator of apoptosis; BCL-2, B-cell lymphoma 2; BECN1, Beclin 1; Bim, BCL2-Like 11; BiP, immunoglobulin heavy-chain-binding protein; B-Raf, B-Raf Proto-Oncogene, Serine/Threonine Kinase; bZIP, basic leucine zipper ;CHOP, CCAAT/enhancer-binding (C/EBP) homologous protein; CNX, calinexin; CRT, calreticulin; DR5, death receptor 5; EGFR, Epidermal Growth Factor Receptor; EIF2AK3, eukaryotic translation initiation factor 2α kinase 3; eIF2, eukaryotic initiation factor 2; EMT, epithelia-tomesenchymal transition; ER, Endoplasmic Reticulum; ERAD, ER-associated degradation; Ero1, ER oxidoreductin 1 Ero1; FOX, Forkhead box; FKBP,FK506‐binding protein; GADD34, Growth arrest and DNA-damageinducible protein 34; GRP58, glucose-regulated protein 58-kD; GRP, glucose regulated protein; GRP78, glucose-regulated protein of 78 kDa; GRP94, glucose regulated protein 94 kDa; GRP170, glucose regulated protein 170; Her2,human epidermal growth factor receptor 2; H-Ras, Harvey Rat Sarcoma Viral Oncogene Homolog; HSPA5, Heat Shock Protein Family A Hsp70 Member 5; IRE1α, Inositol Requiring Enzyme 1α; JNK, Jun N-terminal kinase; LAMP3, lysosomal-associated membrane protein 3; LCN2, lipocalin 2; MAP1LC3B, microtubule-associated protein 1 light chain 3β; MAPK8, Mitogen-Activated Protein Kinase 8; MEF, mouse embryo fibroblast; mTOR, Mechanistic Target Of Rapamycin; Myc, MYC proto-oncogene; PDIs,proteindisulphide isomerases; PERK, protein kinase R (PKR)-like ER kinase; PI3K, Phosphoinositide-3-kinase; p58IPK , Protein Kinase Inhibitor Of 58 KDa; PPI, peptidyl-prolyl isomerase; PUMA, P53 Up-Regulated Modulator Of Apoptosis; RIDD, IRE1α-dependent decay; RIP, regulated intramembrane proteolysis; S1P, site-1 proteases; S2P, site-2 proteases; Sequestosome-1 (SQSTM1); TNF, Tumour necrosis factor; TRAF2, TNF-receptor-associated factor 2; TRIB3, tribbles homologue 3; UGGT, UDP-glucose/glycoprotein glucosyl transferase; uORF , upstream short open reading frame; UPR, unfolded protein response; VCP/p97, Valosin-containing protein; VEGF, Vascular endothelial growth factor; XBP-1,X-box binding protein 1, YY1, Yin Yang 1
.

EP

45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65 66 67 68 69 70 71 72 73 74 75 76 77 78 79 80

Abbreviations:

AC C

44

82

ER stress and unfolded protein response

83

Cells orchestrate a finely-tuned balance between protein synthesis and degradation to

84

maintain protein homeostasis (proteostasis). The endoplasmic reticulum (ER) is a cellular

85

organelle dedicated to the folding and assembly of secretory and membrane-bound proteins

86

and is enriched for enzymes that facilitate the folding process (Anelli and Sitia, 2010). The

87

accumulation of misfolded/unfolded proteins in the ER lumen which causes a condition

88

known as ‘ER stress’ and triggers the unfolded protein response (UPR) to maintain cellular

89

proteostasis by adapting protein synthesis, degradation, trafficking, and folding (Dufey et al.,

2

ACCEPTED MANUSCRIPT 2014). Upon ER stress, molecular sensors in the ER recognize unfolded proteins and relay

91

information across the ER membrane to activate downstream signalling molecules and

92

transcription factors in order to initiate a comprehensive adaptive gene expression program

93

to alleviate ER stress. This gene expression program increases the ER folding capacity by

94

expanding the size of the ER and by increasing the amounts of folding chaperones and

95

modifying enzymes (Ron and Walter, 2007). UPR induction can also enhance the removal

96

of irreparable misfolded proteins from the lumen via ER-associated degradation (ERAD) and

97

autophagy (Rashid et al., 2015). Although UPR induction can promote cell survival in the

98

short term, prolonged UPR activation without the reestablishment of proteostasis leads to

99

cell death primarily by apoptosis.

RI PT

90

SC

100

ER stress and UPR activation are also involved in the pathogenesis and development of

102

many pathological conditions, including diabetes, obesity, neurodegenerative diseases and

103

cancer (Kaushik and Cuervo, 2015). More than 30% of cellular proteins are folded in the ER,

104

which provides a highly specialized microenvironment for protein processing(Anelli and

105

Sitia, 2010). These ER-specific mechanisms are executed by ER-associated chaperones

106

and polypeptide-modifying enzymes, such as glucose regulated proteins (GPRs), calinexin

107

(CNX), calreticulin (CRT), peptidyl-prolyl isomerases (PPIs) and protein-disulphide

108

isomerases (PDIs), Moreover, the ER and UPR components also play key roles in the

109

degradation of irreparably misfolded proteins that need to be disposed of to ensure that only

110

properly folded proteins are exported from the ER (Nakatsukasa and Brodsky, 2008).

TE D

M AN U

101

111

Protein folding: ER chaperones and Foldases

113

In ER, chaperones and folding enzymes operate on the newly synthesized unfolded

114

polypeptides to facilitate their folding into their native conformations and quality control

115

(Braakman and Hebert, 2013). These ER chaperones bind to newly synthesized unfolded

116

proteins, recruit folding enzymes and prepare the unfolded proteins for subsequent folding

117

and assembly. The folding process is also enabled by many folding-enzymes (foldases)

118

which include the PDI family of thiol oxidoreductases and PPIs.

120 121

AC C

119

EP

112

General chaperones

122 123

The general chaperone GRP78/BiP(glucose-regulated protein of 78 kDa/immunoglobulin

124

heavy-chain-binding protein), is one of the most important and abundant ER chaperones,

125

and it recognizes polypeptides that have hydrophobic residues . GRP78/BiP expression is

126

induced by the presence of misfolded or unfolded proteins in the ER (Dufey et al., 2014;

3

ACCEPTED MANUSCRIPT Kozutsumi et al., 1988), and it identifies misfolded proteins and facilitate their folding into

128

their native conformations. Moreover, GRP78/BiP also binds to nascent hydrophobic

129

proteins and protects them from aggregation (Morris et al., 1997). However, if misfolded

130

proteins are persistently resistant to the folding process, they will be targeted to the ERAD

131

machinery for degradation (Chambers et al., 2012; Travers et al., 2000). Recent studies

132

have shown that GRP78/BiP is also important for retro-translocating misfolded proteins to

133

the cytosol for ERAD (Araki and Nagata, 2012; Chillaron and Haas, 2000; Hagiwara et al.,

134

2011). Furthermore, GRP78/BiP also maintains the permeability barrier between the cytosol

135

and the ER lumen by gating the heteromeric Sec61 translocon complex (Plemper et al.,

136

1997). Like GRP78/BiP, the other common general chaperones GRP94 (also as gp96 and

137

HSP90B1), and GRP170 (also as HSP110) also play similar critical roles in facilitating

138

protein folding, assembly and transport as well as the export of misfolded proteins to the

139

cytosol for degradation via the ERAD pathway; however, they have much narrower

140

substrate specificities (Braakman and Hebert, 2013). After the proteins leave the translocon,

141

their folding and post-translational modifications are mediated further by additional

142

collections of chaperones and folding enzymes.

M AN U

SC

RI PT

127

143 Lectin Chaperones

145

The membrane-bound CNX and the soluble CRT are two lectin chaperones that recognize

146

glycoproteins in the ER lumen. These lectins specifically bind mono-glycosylated

147

glycoproteins and are involved in their folding and quality control. When nascent proteins

148

enter the ER lumen, this will activate the ER enzymes glucosidase I and II, which will cleave

149

off excess oligosaccharides to generate the mono-glycosylated proteins so that they can be

150

recognized by CNX and CRT(David et al., 1993; Wada et al., 1995). However, if the protein

151

is still not folded properly, UDP-glucose/glycoprotein glucosyl transferase (UGGT) will act on

152

the unfolded glycoproteins, allowing the proteins to rebind CNX and CRT, and channel them

153

back into the CNX/CRT cycle to undergo more cycles of oxidative folding (Totani et al.,

154

2009). This process will be repeated at least several times to help with protein folding, but

155

persistently misfolded proteins are identified and destroyed via the ERAD pathway.

EP

AC C

156

TE D

144

157

Other Folding Chaperones and Enzymes

158

A key step in the protein folding process is the donation of disulfide bonds to the newly

159

synthesized client proteins and this is mediated by ER resident oxidoreductases, which

160

include ER oxidoreductin 1 (Ero1). Ero1 and the thioredoxin-like PDIs are the major folding

161

enzymes in the ER lumen, which catalyze the formation and breakage of disulfide bonds

162

between cysteine residues within proteins (Frand and Kaiser, 1999). ERp57/GRP58 is

163

another key member of PDI family, which functions to ensure correct folding and the quality

4

ACCEPTED MANUSCRIPT control of newly-synthesized glycoproteins (Oliver et al., 1999) . This role of ERp57/GRP58

165

requires its interaction with the CRT or CNX, which are responsible for recognizing and

166

binding monoglucosylated proteins (Molinari and Helenius, 1999). PPI family of folding

167

enzymes are comprised of Cyclosporin A (CsA)‐binding cyclophilins, the FK506‐binding

168

proteins (FKBPs), and the Parvulin‐like PPIs; these catalyze the conversion of the cis and

169

trans isomers of peptide bonds to proline, and have an essential role in the folding of newly

170

synthesized proteins. In summary, PPIs and PDIs together with other foldases assemble the

171

protein folding process (Jansen et al., 2012).

RI PT

164

172 173

ER stress sensors

SC

174

UPR signalling is activated upon the detection of misfolded proteins in the ER lumen

176

primarily by the chaperone GRP78/BiP and is mediated through three signalling axes driven

177

by three ER-transmembrane sensor proteins, PERK [protein kinase R (PKR)-like ER

178

kinase], IRE1α (Inositol Requiring Enzyme 1α) and ATF6α (Activating Transcription Factor

179

6α), respectively (Hetz, 2012). Under normal unstressed conditions, these ER stress sensor

180

proteins bind GRP78/BiP and remain inactive (Bertolotti et al., 2000; Shen et al., 2002). In

181

response to ER stress, GRP78/BiP binds to the unfolded or misfolded proteins in the ER

182

lumen, and its dissociation from PERK, IRE1α and ATF6 results in their activation and the

183

transduction of downstream UPR signals across the ER membrane to the cytosol and

184

ultimately to the nucleus to trigger an adaptive gene expression program (Bertolotti et al.,

185

2000; Shen et al., 2002)(Figure 1).

186

TE D

M AN U

175

PERK Pathway

188

PERK (eukaryotic translation initiation factor 2α kinase 3, EIF2AK3), is a type I

189

transmembrane kinase with a luminal and a cytoplasmic domain. When active, its key

190

function is to phosphorylate the α subunit of eukaryotic initiation factor 2 (eIF2) on serine 51.

191

This reduces the levels of eIF2-GTP available for translation initiation, resulting in the

192

attenuation of global protein synthesis and thus alleviating the demands for the ER protein

193

folding machinery. (Figure 1).

AC C

EP

187

5

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

Although general translation is decreased in response to ER stress, specific mRNAs are

196

preferentially translated. Activating transcription factor 4 (ATF4) is central to PERK-governed

197

signaling and regulates multiple functions to aid cells recovering from ER stress. For

198

example, under normal unstressed conditions, ATF4 mRNA translation is repressed

199

because its upstream short open reading frame (uORF) is within the 5´- untranslated region

200

of its mRNA, an inhibitory element that blocks ATF4 expression (Harding et al., 2000;

201

Vattem and Wek, 2004). However, the phosphorylation of eIF2α overcomes this inhibition

202

and increases ATF4 protein synthesis. The upregulated ATF4 transcription factor triggers

203

the expression of pro-survival gene products, which include antioxidant response molecules

204

and those involved in protein folding and differentiation (Wang and Kaufman, 2014).

205

Moreover, ATF4 also induces the transcription of CHOP (CEBPz), another key ER stress

206

transcription factor which has both pro-apoptotic and survival functions. In addition, ATF4

207

and CHOP heterodimers also activate the transcription of genes that control autophagy,

208

such as p62/SQSTM1, Atg5 Atg7, and Atg10, to promote cell survival (Huggins et al., 2015;

209

Rouschop et al., 2010). PERK activation also induces the expression of miR-211, which

210

represses CHOP expression and restricts it from activating pro-apoptotic genes (Chitnis et

211

al., 2012). Conversely, ATF4 and CHOP also co-operate to promote the transcription of the

212

GADD34, the regulatory subunit of protein phosphatase 1, which functions as a negative

213

regulator in a feedback loop in PERK signalling to restore protein synthesis. GADD34

214

dephosphorylates

AC C

EP

TE D

194 195

eIF2α,

resulting

in

the

6

eventual

restoration

of

global

mRNA

ACCEPTED MANUSCRIPT translation(Han et al., 2013; Novoa et al., 2003). ATF4 and CHOP also upregulate other

216

gene families that promote protein synthesis, and while this function is essential for the

217

restoration of steady-state protein synthesis in cells with resolved ER stress, it contributes to

218

cell death in cells that cannot rectify the underlying stress (Han et al., 2013). In addition,

219

another ATF4/CHOP target, p58(IPK) can also bind to the PERK kinase domain and inhibit

220

its auto-phosphorylation activity, resulting in a reduction in eIF2α phosphorylation and the

221

synthesis of GRP78/BiP(Yan et al., 2002). Under chronic stress conditions, persistent CHOP

222

induction will also drive cells to undergo apoptosis through promoting the transcription of

223

pro-apoptotic genes, such as the p53-upregulated modulator of apoptosis (also called

224

BBC3), tribbles homologue 3 (TRIB3), the B-cell lymphoma 2 (BCL-2) family of apoptotic

225

regulators, lipocalin 2 (LCN2) and death receptor 5 (DR5 or TNFRSF10B(Hsin et al., 2012;

226

Li et al., 2006; Yamaguchi and Wang, 2004))However, the role of DR5 in ER stress-induced

227

apoptosis has recently been called into question(Glab et al., 2017).

SC

RI PT

215

M AN U

228 229

IRE1α Pathway

230

Upon ER stress, the IRE1α released by the chaperone GRP78/BiP undergoes dimerization,

231

autophosphorylation, and activation, although some argue that misfolded proteins binds to

232

Ire-1 ligands

233

endonuclease activity of IRE1α to splice off a 26-base pair intron from the X-box binding

234

protein 1 (XBP1) mRNA (Yoshida et al., 2001) to generate a specifically spliced XBP1s

235

mRNA species that encodes for the transcriptionally active form of XBP1 (XBP1s). XBP1s

236

drives the expression of genes. chaperones and foldases) that are involved in protein folding

237

as well as ERAD }(Calfon et al., 2002; Lee et al., 2003; Travers et al., 2000). Activated

238

IRE1α also cleaves a range of mRNA species to reduce the protein load in ER, a process

239

named regulated IRE1α-dependent decay (RIDD)(Hollien et al., 2009) (Wang and Kaufman,

240

2016). It has also been demonstrated that activated IRE1α is able to bind to the TNF-

241

receptor-associated factor 2 (TRAF2) and recruit Apoptosis Signal Regulated Kinase 1

242

(ASK1) to an IRE1α-TRAF2-ASK1 complex, which promotes ER stress-induced c-Jun N-

243

terminal kinase (JNK) activation(Urano et al., 2000). Furthermore, the IRE1α-TRAF2

244

complex is also required for the activation of the ER-resident Caspase-12 to facilitate ER

245

stress-induced apoptosis(Yoneda et al., 2001). Another link between the UPR and the

246

apoptotic pathway is the regulation of IRE1α-XBP1 by some members of the Bcl-2 family.

247

For example, the pro-apoptotic Bcl-2-like proteins Bax and Bak are able to bind to the

248

cytosolic domain of IRE1α directly and trigger the downstream signalling (Hetz et al., 2006).

249

Another example is that the BH3-only proapoptotic Bcl-2 family members Bim and PUMA

250

are involved in the regulation of XBP1 splicing via interaction with IRE1α (Rodriguez et al.,

AC C

EP

TE D

in yeast to regulate UPR(Gardner and Walter, 2011) . This triggers the

7

ACCEPTED MANUSCRIPT 2012). Furthermore, it has been shown that RIDD is up-regulated upon ER stress and

252

exhibits pro-apoptotic effects (Maurel et al., 2014). (Figure 2)

253 254 255 256 257

EP

TE D

M AN U

SC

RI PT

251

ATF6 Pathway

259

ATF6 is an ER stress transmembrane protein sensor that functions as a transcription factor

260

upon cleavage. In response to ER stress, the loss of BiP binding releases ATF6 allowing it

261

to translocate from the ER to the Golgi. In the Golgi, ATF6 is cleaved by site-1 (S1P) and

262

site-2 proteases (S2P) through regulated intramembrane proteolysis (RIP) to release a

263

cytosolic amino terminal fragment that functions as an active basic leucine zipper (bZIP)

264

transcription factor, named ATF6f. The cleaved ATF6 fragment migrates to the nucleus to

265

transcriptionally up-regulate protein-folding enzymes and chaperones. GRP78, GRP94,

266

ERp72, and PDI) (Teske et al., 2011) as well as components of ERAD (Shen et al., 2002).

267

For example, upon ER stress, the activated ATF6 relocates to the nucleus to cooperate with

268

the transcription factor Yin Yang (YY1) to transactivate the promoter of the gene (HSPA5)

269

encoding GRP78/BiP, a prosurvival ER chaperone (Baumeister et al., 2005).

AC C

258

8

ACCEPTED MANUSCRIPT 270

ATF6 also plays a pivotal role in transcriptional induction of other critical ER-stress

271

transcription factors, such as XBP1 (Lee et al., 2002; Yoshida et al., 2001; Yoshida et al.,

272

2000) and CHOP (Nakanishi et al., 2005; Yoshida et al., 2000). Accordingly, XBP1 mRNA

273

expression is induced by ATF6 and spliced by IRE1 to be activated in response to ER

274

stress. In addition, ATF6 also cooperates with XBP1, to upregulate genes, encoding ER

275

protein chaperones, ERAD signalling complexes, and lipid biosynthetic enzymes.

277

ER stress and Cancer

278

ER Chaperones and Cancer regulation:

RI PT

276

279

The high rates of translation associated with cancer cell proliferation and metabolism can

281

induce ER stress and consequently UPR (Ozcan et al., 2008). In addition, large-scale gains

282

and losses of genetic material as well as point mutations generate imbalanced protein levels

283

and proteins that are difficult to fold, thereby challenging proteostasis-maintaining

284

mechanisms in cancer cells (Deshaies, 2014). In spite of the precarious proteostatic balance

285

in cancer cells there is considerable evidence to support the notion that the UPR has a

286

central role in cancer initiation, progression and chemotherapeutic resistance (Wang and

287

Kaufman, 2014). For example, GRP78/BiP overexpression is commonly observed in

288

cancers and is associated with aggressive cancer growth and metastasis (Miao et al., 2013).

289

Similar observations have been made for other proteostasis-promoting molecules, including

290

the ubiquitously expressed protein degradation mediator VCP/p97 (Yamamoto et al., 2003).

291

TE D

M AN U

SC

280

ER sensors and Cancer:

293

Oncogenic stress is increasingly recognized as a cause of ER stress and a UPR trigger.

294

Oncogene activation, such as B-Raf proto-oncogene mutations, H-Ras proto-oncogene

295

mutations, and c-Myc amplification, as well as chemotherapeutic drug treatments, can

296

induce ER stress in cancer cells (Bu and Diehl, 2016). For example, expression of the c-Myc

297

oncogene can increase general transcription and translation, which causes the production of

298

high levels of misfolded and unfolded proteins in the ER lumen. As a result, PERK will be

299

induced to activate survival mechanisms, such as autophagy, which can promote tumor

300

initiation and progression(Hart et al., 2012). This has been observed in both human

301

lymphoma cells and mouse models carrying c-Myc translocations (Hart et al., 2012).

302

Another oncogenic molecule involved in inducing ER stress is B-RAF, which plays an

303

important role in the development of melanoma. Mutant B-RAF can induce chronic ER

304

stress via the binding to BiP in the ER lumen, and this

305

GRP78/BiP from the three ER sensor arms. As a result, PERK and IRE1α are activated to

AC C

EP

292

9

leads to the dissociation of

ACCEPTED MANUSCRIPT 306

promote autophagy to promote cell survival(Corazzari et al., 2015)

307

adenocarcinoma, another oncogene HER2/Neu, has also been found to activate PERK

308

signalling and thereby, cancer progression(Bobrovnikova-Marjon et al., 2010). Furthermore,

309

PERK activation also helps cancer cells to overcome environmental stress, such as hypoxia

310

and a lack of nutrition. For instance, at low glucose levels, PERK activation induces cancer

311

cell survival via Akt activation and hexokinase II transportation to the mitochondria(Hou et

312

al., 2015). The UPR is also intimately involved in angiogenesis, a tumor progression process

313

that facilitates the enrichment of cancer cells with oxygen and nutrition’s via the expansion

314

of the vascular network. For example, VEGF, a proangiogenic factor, has been found to

315

induce cell survival and angiogenesis in endothelial cells through activating PERK signalling

316

and ATF6. Furthermore, PERK-ATF4 arm has also been shown to repress anti-

317

angiogenesis factors and enhance Vascular endothelial growth factor VEGF expression

318

(Blais et al., 2006). In addition, the PERK/elF2α arm and its downstream molecules also

319

have a broad role in the regulation of autophagy to promote cancer cell survival. In human

320

cancer cells, hypoxia can upregulate PERK to induce the expression of autophagy

321

molecules ATG5 and microtubule-associated protein 1 light chain 3β (MAP1LC3B), which

322

are responsible for the expansion of phagophores. Conversely, PERK inhibition causes the

323

downregulation of both MAP1LC3B and ATG5, further suggesting a role of PERK in

324

promoting autophagy and thus cancer development (Rouschop et al., 2010). In addition, the

325

oncogene c-Myc can also regulate PERK to activate the cytoprotective autophagy

326

mechanism in cancer cells (Yang et al., 2013). The PERK/elF2-α/ATF4 signalling axis has

327

been shown to be involved in autophagy activation in neural cells after ionic irradiation by

328

increasing the expression of autophagic regulator LC3. In agreement, PERK-knockdown

329

neural cells showed enhanced sensitivity to radiation (Yang et al., 2013).

EP

TE D

M AN U

SC

RI PT

Moreover, in

330

The transcription factors ATF4 and CHOP are downstream targets of PERK and have been

332

found to be involved in autophagy regulation and autophagosome formation. The PERK-

333

elF2α-ATF4 signalling axis regulates the expression of p62/SQSTM1, a regulator and

334

established genetic marker of autophagy, in response to leucine starvation in mouse embryo

335

fibroblast (MEFs), p62/SQSTM1 also enhances stem-like properties by stabilizing MYC

336

mRNA in breast cancer. Moreover, CHOP is one of the transcription factors that induces

337

p62/SQSTM1 expression, and appropriately, CHOP knockdown restricts p62/SQSTM1

338

expression and autophagy (B'Chir et al., 2013). Furthermore, apart from PERK activation,

339

ATF4 also controls LC3B expression in response to the bortezomib (proteasome inhibitor)

340

treatment, inducing autophagy and drug resistance in the breast carcinoma cells (Milani et

341

al., 2009).

AC C

331

10

ACCEPTED MANUSCRIPT 342 PERK signalling also plays an important role in cancer metastasis and migration in hypoxic

344

microenvironments. ATF4 stimulates the expression of the lysosomal-associated membrane

345

protein 3 (LAMP3) to promote epithelia-to-mesenchymal transition (EMT) and metastatic

346

distribution in several cancer types (Pytel et al., 2016). In conclusion, PERK signalling plays

347

a crucial role in regulating cell fate in response to internal and external stresses; as such,

348

PERK might help cancer cells to adapt to stress conditions and activate the survival

349

pathway,

350

chemotherapeutic drug resistance.

such

as

autophagy,

angiogenesis,

cell

cycle

351

RI PT

343

arrest,

metastasis

and

In human prostate cancer cells, IRE1α regulates cyclin A1 activity and increases cell

353

proliferation via the control of XBP1 splicing (Thorpe and Schwarze, 2009). XBP1, a key

354

transcription factor of the UPR, is essential for the survival of cancer cells under hypoxia and

355

for tumor growth (Romero-Ramirez et al., 2004). For instance, human lung carcinoma and

356

fibrosarcoma cells increase XBP1 splicing and GRP78/BiP expression in response to

357

hypoxic stress (Vandewynckel et al., 2013). In addition, XBP1 expression and splicing were

358

also up-regulated in breast cancer as well as hepatocellular carcinoma (HCC), which may

359

promote cell survival via the expression of GRP78/BiP (Fujimoto et al., 2003; Shuda et al.,

360

2003). Furthermore, XBP1 overexpression is necessary for plasma cell differentiation and

361

expansion, which may be important for the initiation of multiple myeloma (Carrasco et al.,

362

2007). Nutrient deprivation is also one of the tumor microenvironment factors that trigger ER

363

stress and thereby UPR. XBP1 activity is also associated with the stress response towards

364

glucose deprivation, where glucose deprivation enhances XBP1 splicing (Spiotto et al.,

365

2009). VEGF is an essential regulator of tumor angiogenesis and therefore expansion. UPR

366

promotes

367

accordingly, depletion of either IRE1α or ATF6 causes downregulation of VEGF expression

368

and in vivo angiogenesis in tumor neovascularization mouse models (Ghosh et al., 2010; Liu

369

et al., 2013).

M AN U

TE D

EP

angiogenesis

through

IRE1α and

ATF6-mediated

VEGF

induction,

and

AC C

370

SC

352

371

The cross talk between ER stress and Autophagy in Cancer:

372

Autophagy is an important means by which tumor cells defend against microenvironmental

373

stress and chemotherapeutic drugs. ER stress is intimately involved in macro-autophagy

374

activation and autophagosomal membrane formation (Hart et al., 2012). UPR regulates

375

autophagy through the IRE1α, the PERK/elF2α and the intracellular calcium-dependent

376

signalling axes (Hart et al., 2012; Høyer-Hansen and Jäättelä, 2007) via three key cellular

377

signalling molecules: Jun N-terminal kinase (JNK), AKT1 and mTOR. IRE1α can activate

11

ACCEPTED MANUSCRIPT JNK (also called MAPK8; Mitogen-activated protein kinase 8), which has the ability to

379

phosphorylate anti-apoptotic protein BCL-2 and inhibit its interaction with BECN1, a

380

molecule responsible for the activation of PtdIns3K and phagophore formation in autophagy

381

(Clarke et al., 2012; Strappazzon et al., 2011). Moreover, the post-transcriptional splicing of

382

XBP1 mRNA by IRE1α gives rise to the transcription factor XBP1s, which induces the

383

expression of BCL-2. Furthermore, XBP1s can also transcriptionally activate the expression

384

of BECN1 directly (Rashid et al., 2015).

385

The PERK/elF2α arm and its downstream signalling molecules play a crucial role in the

386

regulation of autophagy. In human cancer cells, PERK induces the expression of the

387

autophagy molecules, ATG5 and MAP1LC3B, which are responsible for the expansion of

388

phagophore (Rouschop et al., 2010). The PERK/elF2α/ATF4 arm is also involved in

389

autophagy activation by increasing the expression of LC3 in nerve cells in response to

390

radiotherapy (Yang et al., 2013).

391

ATF4 and CHOP are downstream target molecules of PERK, which has been found to be

392

involved in autophagy regulation and autophagosomal formation. elF2α/ATF4 regulate p62,

393

a marker gene and regulator of autophagy, after inducing leucine starvation in mouse

394

embryo fibroblasts (MEFs). Moreover, CHOP is one of the transcription factors that induces

395

p62 expression, and accordingly, knockdown of CHOP causes the depletion of p62 and the

396

repression of autophagy (B'Chir et al., 2013). Furthermore, apart from PERK activation,

397

ATF4 also regulates LC3B in response to the Bortezomib treatment in the breast cancer

398

MCF-7 cells to induce autophagy and resist the cytotoxic effects of bortezomib (Milani et al.,

399

2009). The ATF6 signalling arm plays a crucial role in mediating autophagy. In collaboration

400

with CHOP, ATF6 inhibits the expression of death-associated protein kinase 1 (DAPK1), an

401

activator of IFNγ-induced cell death, to promote autophagy (Kalvakolanu and Gade, 2012).

402

Moreover, it is well established that ATF6 transcriptionally activates BiP/GRP78, which

403

indirectly represses mTOR through AKT1 downregulation (Yung et al., 2011). However,

404

GPR78 knockout in mice caused the activation of AKT, which suggests that GRP78 has two

405

different regulatory roles in the regulation of autophagy (Chang et al., 2012).

406

UPR can regulate autophagy indirectly through molecules that stimulate or inhibit mTOR.

407

For example, TSC1/2 (tuberous sclerosis) is a negative regulator of mTOR. Indeed,

408

research showed that the induction of ER stress in TSC-deficient MEF cells did not

409

downregulate mTOR activity (Qin et al., 2010). Another ER stress modulator and

410

transcription factor regulated by CHOP and ATF4 is TRIB3, which is a negative regulator for

411

AKT-mTOR, which causes autophagy activation (Verfaillie et al., 2010).

AC C

EP

TE D

M AN U

SC

RI PT

378

412 413

The relationship between FOXO, ER stress and cancer

12

ACCEPTED MANUSCRIPT The FOXO subgroup of Forkhead box (FOX) transcription factors were first identified in the

415

nematode Caenorhabditis elegans (C. elegans) as Daf-16 for its role in metabolic signalling

416

and longevity (van der Horst et al., 2006). In mammals, there are four FOXO proteins:

417

FOXO1, FOXO3, FOXO4, and FOXO6. Structurally, they are all composed of a highly

418

conserved DNA binding domain, located upstream of a nuclear localization signal, a nuclear

419

export sequence and a C-terminal transactivation domain (Anderson et al., 1998). Amongst

420

all FOXO proteins, FOXO3 is the most ubiquitously expressed within both embryonic and

421

adult tissues (Furuyama et al., 2000; Greer and Brunet, 2005). FOXO3 knock-down results

422

in premature ovarian failure, spontaneous lympho-proliferation, organ inflammation, and

423

helper T cell hyperactivation (Lin et al., 2004; Paik et al., 2007; Tothova et al., 2007). The

424

relatively mild and tissue-restricted phenotype observed in FOXO3-deficient animals

425

indicates functional and expression compensation exists between these different FOXO

426

proteins. FOXO activity is tightly regulated but is also fine-tuned by a range of post-

427

translational modifications, including phosphorylation, ubiquitination, acetylation and

428

methylation (Calnan and Brunet, 2008; Lam et al., 2013). The most extensively researched

429

post-translational modification is Akt(PKB)-mediated phosphorylation, which promotes

430

FOXO3 cytoplasmic translocation and thereby inhibition in response to growth factor

431

signalling (Lam et al., 2013; Myatt and Lam, 2007). By contrast, phosphorylation by AMP-

432

activated protein kinase (AMPK)(Greer et al., 2007), p38-MAPK (Consolaro et al., 2015; Ho

433

et al., 2012), JNK (Sunters et al., 2006) promotes nuclear localization and the activation of

434

FOXO protein activation. For example, JNK recognizes FOXO3 through at least 4

435

acetylation sites (K242, K259, K290 and K569) to mediate its phosphorylation. The JNK-

436

dependent S574 phosphorylated form of FOXO3 has been shown to be involved in the

437

induction of apoptosis (Li et al., 2016). FOXO proteins function by binding to the promoter

438

consensus sequences 5’-GTAAA(T/C)A-3’, and modulating the transcription of target genes

439

through their transactivation domain and interaction with a myriad of co-factors (Greer and

440

Brunet, 2005; Lam et al., 2013). Amongst their many functions, FOXO proteins can

441

negatively regulate cell cycle progression through activating negative cell cycle regulators,

442

including p21Cip1, p27Kip1, and p130(RB2). In addition, FOXO is also involved in the

443

regulation of programmed cell death through the transcriptional control of proapoptotic

444

genes, such as tumor necrosis factor (TNF), and the Bcl-2 related Bim and PUMA (Lam et

445

al., 2013). Moreover, the cytostatic and cytotoxic activity of common cancer therapeutic

446

agents, such as anthracyclines, taxanes and platinum compounds, also relies on FOXO3

447

activity (Koo et al., 2012). One of the downstream target genes repressed by FOXOs is the

448

potent oncogene FOXM1. FOXM1 is another member of the FOX family of transcription

449

factors that is widely expressed in actively proliferating tissues and plays a key role in

450

tumorigenesis and cancer progression (Bella et al., 2014; Koo et al., 2012; Lam et al., 2013;

AC C

EP

TE D

M AN U

SC

RI PT

414

13

ACCEPTED MANUSCRIPT Zona et al., 2014). FOXO3 and FOXM1 transcription factors regulate the expression of a

452

common group of genes, but have antagonistic functions in regulating their targets. FOXO3

453

and FOXM1 have also been shown to negatively regulate one another’s expression and

454

activity, and appropriately, FOXM1 is described as an antagonist of FOXO proteins (Fu and

455

Tindall, 2008). Recent evidence also suggests FOXM1 can protect cells from

456

chemotherapeutic drug-induced senescence and cell death, and appropriately, FOXM1 is

457

overexpressed in genotoxic and cytotoxic agent-resistant cancer cells (Karunarathna et al.,

458

2016; Khongkow et al., 2015; Khongkow et al., 2014; Kwok et al., 2010; Monteiro et al.,

459

2013; Myatt et al., 2014). Collectively, these findings illustrate that FOXO proteins, and

460

particularly FOXO3, have a crucial tumor suppressive role (Lam et al., 2013).

461

PERK pathway and FOXO3 story:

462

FOXO3 is an integral molecule at the heart of ER-stress and UPR signalling. PERK, one of

463

the three most important ER stress sensors, has been shown to be able to modulate

464

FOXO3 activity both directly and indirectly. PERK, one of the main signalling arms of UPR,

465

has also been found to induce FOXO transcription factor activity to promote insulin

466

resistance (Zhang et al., 2013). (Figure 3).

467 468

The PERK pathway plays a key role in insulin resistance, obesity and adipocytic

469

differentiation. In these conditions, PERK can cause mTOR induction as well as AKT

470

phosphorylation on Ser473 and activation (Bobrovnikova-Marjon et al., 2012). Thus, upon

471

ER stress and PERK activation, Akt can phosphorylate FOXO proteins and cause them to

472

relocate from the nucleus to the cytoplasm where FOXOs are sequester by head shock

AC C

EP

TE D

M AN U

SC

RI PT

451

14

ACCEPTED MANUSCRIPT proteins and become inactivated (Zhang et al., 2015). In consequence, constitutive PERK

474

activation can negatively regulate FOXO activity indirectly via Akt to restrict apoptotic

475

signalling and thus, promote cancer progression. Paradoxically, PERK can also mediate

476

FOXO3 phosphorylation directly, in an Akt-independent manner, at specific serine residues,

477

such as S261, S298, S301, S303 and S311 to promote FOXO3 its nuclear relocation and

478

activation(Zhang et al., 2013). In addition, downstream of PERK, CHOP-induced

479

enhancement of ERO1α expression can also trigger ROS production and thereby, the

480

induction of the JNK. This can, in turn, phosphorylate and activate FOXO proteins.

481

Furthermore, in cells treated with the ER-stress inducing agent tunicamycin, CHOP

482

cooperates with FOXO3 to induce the expression of proapoptotic genes, including Bim and

483

PUMA in response to ER stress (Ghosh et al., 2012). Nevertheless, the mechanism that

484

dictates whether PERK signalling activates or restrains FOXO3 activity remains enigmatic.

485

Interestingly, recent work on B-Raf-induced melanoma has demonstrated that PERK is a

486

gene dose-dependent tumor suppressor, where the level of its activity determines whether it

487

functions as a tumor suppressor or an oncogene (Pytel et al., 2016). The finding that the

488

nature of PERK function is determined by gene dose and possibly the duration of its activity

489

might help to explain the dual contradictory roles of PERK signalling on FOXO3 regulation.

M AN U

SC

RI PT

473

490 IRE-1 and FOXO regulation:

492

Another possible means by which ER stress may impact on FOXO activity is through the

493

other key ER stress sensor IRE1α. A recent study has shown that the C. elegans FOXO

494

transcription factor Daf-16 and its human homologue FOXO3 can restore proteostasis in Ire-

495

1 mutant and cause ER stress resistance in the nematode C. elegans (Safra et al., 2014).

496

This suggest that FOXO transcription factors can maintain protein homeostasis and cause

497

drug resistance even if the IRE1α-mediated ERAD system is defective and that FOXO

498

proteins function downstream of IRE1α in ER stress signalling. Moreover, XBP1u, which is

499

targeted by IRE1α upon ER stress, can interact with FOXO1 to promote its degradation

500

through 20S proteasome pathway in cancer cells (Zhao et al., 2013). Furthermore, XBP1s

501

also interacts with FOXO1 to direct it toward proteasome-mediated degradation to prevent

502

insulin resistance in type 2 diabetes (Zhou et al., 2011). Moreover, activation of IRE1α can

503

also stimulate JNK (Nishitoh et al., 2002), which has been shown to augment FOXO3

504

activity (Sunters et al., 2003; Sunters et al., 2006). Consistently, recent research also

505

demonstrates that in response to elevated ROS (reactive oxygen species) levels, JNK-

506

mediated

507

cells. Accordingly, treatment of HMSCs with ROS-inducing agent H2O2 leads to JNK-

508

mediated FOXO3 phosphorylation at Ser294 and nuclear translocation to transactivate

AC C

EP

TE D

491

FOXO3 induces

autophagy

in

15

human

mesenchymal

stem

(HMSCs)

ACCEPTED MANUSCRIPT 509

autophagy genes (Gomez-Puerto et al., 2016). Collectively, these findings propose a key

510

role of FOXO proteins in linking ER stress to cancer development as well as cancer

511

microenvironmental and cytotoxic stress resistance.

512 513 Chaperones and FOX regulation:

515

Furthermore, in colorectal cancer, FOXM1 can bind to promoter of GRP78/BiP and

516

regulated its expression at the transcriptional level to increase the levels of chaperone

517

protein and cope with ER stress induced by the accumulation of unfolded proteins

518

(Baumann et al., 2016). Conversely, the ability of FOXM1 to promote invasion and migration

519

of colorectal cancer cells depends on GRP78/BiP (Luo et al., 2016).

520

downregulation of GRP78/BiP reduces the ability of FOXM1 to induce colorectal cancer cell

521

migration and invasion (Luo et al., 2016). In agreement, Heat shock proteins Hsp70 has also

522

been shown to be able to regulate the

523

indirectly by promoting their maturation(Colvin et al., 2014). This indicates the existence of a

524

feed forward signalling loop involving FOXM1 and GRP78/BiP in the regulation of cancer

525

progression.

526

ER stress and FOX regulation in Worms:

527

The nematode worm Caenorhabditis elegans and fruit fly Drosophila melanogaster share

528

numerous diverse biological processes with mammals, which include the highly conserved

529

ER stress and UPR signalling cascades. The C. elegans abnormal dauer formation-16 (Daf-

530

16) and Drosophila dFoxO are sole orthologues of mammalian FOXO transcription factors,

531

including FOXO1, FOXO3 FOXO4 and FOXO6 (Greer and Brunet, 2005). Our

532

understanding of the contribution of the FOXO family of transcription factors in the ER stress

533

response has been accelerated by studies in worm and fly models. Various C. elegans and

534

D. melanogaster ER stress and UPR-related genes have structural orthologues in humans,

535

and these nematode worms and fly orthologues also share their functional relationships with

536

FOXO proteins as their mammalian counterparts (summarized in Table 1 and Table 2,

537 538

respectively).

RI PT

514

SC

Accordingly,

AC C

EP

TE D

M AN U

expression of oncoproteins, such as FOXM1,

C elegans orthologues pek-1

Human gene

Function

eukaryotic translation initiation factor 2-alpha kinase 3 (EIF2AK3), also known as PERK kinase

pek-1 encodes a predicted ncodes a predicted transmembrane protein kinase Eif2ak3 which is required for the UPR. Pek-1 functions in the ER to phosphorylate eIF2α which inhibits assembly of 80S ribosomes and subsequent protein translation initiation, thus counteract endogenous ER stress by reducing the load of proteins to be processed in the ER (Safra et al., 2013; Shen et al., 2001; Shen et al., 2005).

16

Relationship with daf-16/FOXO in C. elegans Pek-1 regulates genes that are involved in inducible-UPR (i-UPR) (Shen et al., 2005). Pek-1 (PERK) modulates Daf-16/FOXO activity and phosphorylates the human and fly orthologues (Wang and Kaufman, 2014). It acts through Ire-1/Xbp-1, which cooperate with Daf-16/FOXO to maintain ER homeostasis (Shen et al., 2001).

ACCEPTED MANUSCRIPT endoplasmic 1 is a transmembrane serine/threonine protein kinase and sitereticulum-tospecific endoribonuclease which is required for UPR that nucleus signaling 1 counteracts cellular stress induced by accumulation of (ERN1), also unfolded proteins in the ER. Upon ER stress, activated IRE-1 known as removes an intron from X-box binding protein-1 (xbp-1) ribonuclease mRNA through unconventional splicing to produce inositol-requiring transcriptionally active Xbp-1(Calfon et al., 2002; Shen et al., protein-1 (IRE-1) 2001; Shen et al., 2005; Urano et al., 2002). The ire-1/xbp-1 pathway regulates the majority of genes in both i-UPR and cUPR, including those that are implicated in protein folding and ERAD. The ire-1/xbp-1 pathway has broader functions and plays a more important role in ER homeostasis than that of pek-1 and atf-6 when compared to mammalian cells (Henis-Korenblit et al., 2010; Shen et al., 2005). XBP-1 It is a bZIP transcription factor which plays an important role in maintaining ER homeostasis and promoting normal development via regulation by ire-1 as mentioned above (Kaufman, 1999; Shen et al., 2001; Shen et al., 2005; Urano et al., 2002). Cullin-1 (CUL-1) Encodes a cullin which is an core component of the SCF (Skp1p, Cullin, and F-box) ubiquitin-ligase complex (E3) that interact directly with various Skr proteins to facilitate ubiquitin-mediated protein degradation via autophagy (Ghazi et al., 2007; Safra et al., 2014). Cullin-2 (CUL-2) Encodes an E3 ubiquitin ligase that regulates diverse biological processes via ubiquitin-mediated proteolysis. CUL-2 functions as part of a Cul2-RING ubiquitin-ligase complex that regulates cell cycle progression, such as the initiation of meiotic anaphase II, cytokinesis and mitotic chromosome segregation (Feng et al., 1999; Sonneville and Gonczy, 2004). Targets of CUL-2-mediated degradation include CYB-1/cyclin B and the zinc-finger protein TRA-1(Liu et al., 2004). SKP1-CUL1-F-box All three genes encode proteins that are core components of (SCF) protein E3 the E3-ubiquitin ligase complex that facilitates ubiquitinubiquitin ligase mediated protein degradation via autophagy. In particular, these proteins are shown to interact with CUL1, C. elegans cullin homolog (Ghazi et al., 2007; Nayak et al., 2002; Yamanaka et al., 2002). SCF protein E3 Functions within the E3 complex to maintain ER ubiquitin ligase homeostasis(Liu et al., 2011; Safra et al., 2014)

xbp-1

skr-2, skr-8, skr-10

skr-5

TE D

cul-2

M AN U

SC

cul-1

RNF-121

hrd-1/sel-11

ERAD-associated E3 ubiquitin-protein ligase (HRD) -1

RNF-121 is a RING finger E3 ubiquitin ligase localized to membranes of the ER, the sarcoplasmic reticulum, and the Golgi. It exhibits E3 ubiquitin-protein ligase activity in vitro and in vivo and functions in ERAD pathway downstream of pek-1(Darom et al., 2010). Encodes an E3 ubiquitin ligase that has similar function as the E3 ubiquitin ligase HRD-1 in human. Hrd1/sel-11, together with its substrate receptor Hrd3/Sel-1, are core components of a large multi-subunit ER-embedded ubiquitin ligase complex that mediate the ubiquitylation of misfolded proteins from the ER lumen as they are retro-translocated out of the ER (Munoz-Lobato et al., 2014; Sasagawa et al., 2007). The gene encodes the Derlin-1 protein, also known as degradation in endoplasmic reticulum protein 1 (DERL1), is part of a complex that includes VIMP, SEL1, HRD1, and HERP. Derlin-1 localizes to endosomes and to the ER and mediates ERAD that detects misfolded proteins in the ER and targets them for degradation (Dang et al., 2011; Mehnert et al., 2014; Schaheen et al., 2009).

AC C

EP

rnf-121

cup-2

Ire-1 acts upstream of Xbp-1, which cooperates with Daf-16/FOXO to modulate ER stress response (Lee et al., 2015; Shen et al., 2001).

RI PT

ire-1

Derlin-1 (DERL1)

cdc-48.1 and cdc48.2

ATP-driven chaperone valosincontaining protein (VCP)/p97

hsp-3 and hsp-4

glucose regulated protein 78/immunoglobulin

The two genes encode a type II AAA ATPase which is an essential component in ERAD. It functions as an ubiquitinselective chaperone which target unfolded proteins for dislocation across the ER membrane, and subsequent degradation by 26S proteasome in the cytosol (Janiesch et al., 2007; Meyer et al., 2012; Nishikori et al., 2008). Encodes a major ER chaperone protein which is involved in many cellular processes, including translocating newly synthesized polypeptides across the ER membrane,

17

Downstream of Ire-1, Xbp-1 collaborates with daf-16/FOXO to activate genes to promote ER stress resistance (Henis-Korenblit et al., 2010). The isoform of Cul-1, which acts upstream of Daf-16/FOXO to promote its nuclear accumulation and transcriptional activity including skr genes. The skr genes encode Skr proteins that are co-factors of Cul-1 (Ghazi et al., 2007).

As part of the SCF protein complex, these genes function upstream of Daf16/FOXO to promote transcriptional activity of Daf-16/FOXO by facilitating its nuclear accumulation (Ghazi et al., 2007). Transcriptional target of Daf-16/FOXO and its regulation is independent of the Ire-1/Xbp-1 pathway (Liu et al., 2011; Safra et al., 2014). Downstream of Pek-1 and Daf16/FOXO signalling (Darom et al., 2010).

Downstream targets of Ire-1/Xbp-1 and Daf-16/FOXO pathway (Safra et al., 2014).

Downstream of Ire-1/Xbp-1 pathway and likely a target of Daf-16/FOXO which is a cofactor of Xbp-1 in ER stress response (Safra et al., 2014).

Downstream of the Ire-1/Xbp-1 and Atf6 pathways which are also the downstream targets of daf-16/FOXO (Caruso et al., 2008; Safra et al., 2014). Downstream of the Ire-1/Xbp-1 pathway, as well as Daf-16/FOXO, and are both up-regulated in response to

ACCEPTED MANUSCRIPT

endoplasmic reticulum oxidoreductase-1, ERO-1

sir-2.1

Sirtuin 1 (SIRT-1)

Upstream activator of Pek-1(Harding et al., 2003).

Upstream activator of Daf-16/FOXO in the insulin-like signaling pathway (Tissenbaum and Guarente, 2001; Wolff and Dillin, 2006).

SC

M AN U

553 Human gene

PERK

Function

Relationship with dFOXO/FOXO in D. melanogaster

It encodes a kinase that phosphorylates dFOXO at S243, promoting its nuclear localization and activity, enhancing the ER stress response. Interestingly, this contrasts the effect of dFOXO phosphorylation by Akt, which results in its sequestration in the cytoplasm (Zhang et al., 2013). It regulates the expression of heat shock proteins (Hsps) at transcription level (Fernandes et al., 1995).

The kinase phosphorylates dFOXO and promotes transcription of downstream ER stress-related genes (Zhang et al., 2013).

AC C

EP

D. melanogaster orthologues perk

554 555 556 557

ER stress (Kapulkin et al., 2005; Urano et al., 2002).

Table 1 Functional human orthologues of C. elegans ER stress-related genes and their functions and relationship with Daf-16/FOXO. A role in the regulations of ER stress response to promote ER stress resistance and longevity. In C. elegans, ER stress responses, particularly the unfolded protein response (UPR), are mainly dependent on the Ire-1/Xbp-1 pathway. In fact, the downstream gene targets of the ire-1/xbp-1 pathway accounts for the majority of UPR genes which are implicated in protein folding and endoplasmic reticulum-associated protein degradation (ERAD), such as cul-1, cul-2, skr, and cup-2. Interestingly, xbp-1 was found to collaborate with daf-16 to activate transcription of these downstream targets. Studies of C. elegans also showed that pek-1 acts synergistically with atf-6 to maintain ER homeostasis as an alternative mechanism to the ire1/xbp-1 pathway. Furthermore, pek-1 and atf-6 are both found to interact with daf-16 during ER stress response, suggesting a role of daf-16 in maintain ER homeostasis that is independent of the ire-1/xbp-1 signaling pathway.

TE D

539 540 541 542 543 544 545 546 547 548 549 550 551 552

ero-1

facilitating folding and assembly of proteins, targeting misfolded proteins for ERAD and acting as an ER stress sensor (Calfon et al., 2002; Lee, 2005). Both proteins have compensatory functions for each other (Kapulkin et al., 2005) It is an ER-localized enzyme that is required specifically for protein oxidation in the ER lumen (Harding et al., 2003). This enzyme mediates disulfide bond formation in the ER by re-oxidizing P4HB/PDI, the enzyme catalyzing protein disulfide formation, which results in the production of reactive oxygen species (ROS) in the cell. Therefore, it plays an important role in ER stress-induced, CHOPdependent apoptosis by activating the inositol 1,4,5trisphosphate receptor (IP3R1) (Sevier and Kaiser, 2008). A histone deacetylase (HDAC) that is known to negatively regulate ER stress response by deacetylating XBP-1, thus inhibiting its transcriptional activity to promote ER stressinduced apoptosis (Tissenbaum and Guarente, 2001; Wang et al., 2011). SIRT1 is also implicated in the regulation of eIF2α phosphorylation which is upstream of CHOP and GADD34, important mediators of stress-induced response(Ghosh et al., 2011).

RI PT

heavy chainbinding protein, (grp78/BiP)

Heat shock transcription factor (hsf)

HSF

hsp70

HSP70

It encodes a protein with ATP-dependent chaperone activity which directs the re-folding of misfolded proteins, thus acts against ER stress (Hartl and Hayer-Hartl, 2002).

Upon induction of ER stress, dFOXO binds to the promoter region of HSF to up-regulate transcription of target genes (Donovan and Marr, 2016). Direct downstream transcription target of HSF as well as dFOXO/FOXO as HSF transcription is regulated by dFOXO/FOXO (Donovan and Marr, 2016; Fernandes et al., 1995).

Table 2. Functional human orthologues of D. melanogaster ER stress-related genes and their functions and relationship with dFOXO/ FOXO. Similarly, in D. melanogaster, dFOXO is shown to regulate ER homeostasis at the transcriptional level by binding to promoter regions of ER stress related genes

18

ACCEPTED MANUSCRIPT Daf-16 and dFOXO and regulation of Ire-1 arm:

559

Appropriately, emerging evidence has revealed that the orthologues of human FOXO

560

proteins, Daf-16 and dFOXO are implicated in the regulation of ER homeostasis at the

561

transcriptional level in both nematodes and flies, respectively. In C. elegans, ER stress

562

responses, particularly UPR, are dependent largely on the Ire-1/Xbp-1 pathway. In fact, the

563

downstream gene targets of the Ire-1/Xbp-1 pathway account for the majority of UPR genes

564

which are implicated in protein folding and ERAD, including cul-1, cul-2, sel-1, sel-2 and

565

cup-2. Interestingly, Daf-16/FOXO also activate transcription of these ER stress downstream

566

targets upon ER stress (Ghazi et al., 2007), suggesting Daf-16/FOXO and Xbp-1 cooperate

567

to regulate genes important for resisting ER stress. Consistently, Xbp-1 has been found to

568

function synergistically with Daf-16 to enhance transcription of ER-stress resistance genes,

569

as well as specific longevity-promoting genes, such as dox-1 (Henis-Korenblit et al., 2010).

570

In C. elegans, activation of the IGFR homologue, Daf-2, stimulates the PI3K-Akt pathway,

571

which will, in turn, phosphorylate Daf-16, promoting its sequestration in the cytoplasm, and

572

thus, restraining its transcriptional activity (Murphy et al., 2003). Studies into long-lived daf-2

573

mutants, where Daf-16 is constitutively activated, have revealed the many diverse roles of

574

Daf-16/FOXO in regulating downstream target genes in the response to ER stress (Lin et

575

al., 2001; Ogg et al., 1997; Tullet et al., 2008). In these daf-2 mutants, the ER stress-

576

inducing agent tunicamycin can induce increased load of misfolded proteins in the ER and

577

activate an arm of the UPR, involving the endoribonuclease Ire-1, and its downstream target

578

Xbp-1. Daf-16/FOXO has also been found to be able to act independently of the Ire-1/Xbp-1

579

pathway in ire-1 and/or xbp-1-deficient animals (Safra et al., 2014). Accordingly, activated

580

Daf-16 has been shown to employ an alternative pathway for the degradation of misfolded

581

ER proteins in the situations of a deleted ire-1 or xbp-1, via transcriptional activation of skr

582

genes, including skr-5 (Safra et al., 2014). These skr gene products form a core component

583

of an E3-Ubiqiutin ligase complex, that mediates degradation of unfolded proteins under

584

conditions of ER stress (Safra et al., 2014). This suggests that upon ER stress the activated

585

Daf-16/FOXO can enable the autophagy-mediated ERAD as well as the proteasome-

586

mediated ERAD to resolve ER protein homeostasis, and that Daf-16 can circumvent the

587

need for Ire-1/Xbp1 for ER homeostasis and function. Interestingly, Skr proteins can also

588

interact directly with Cul-1, another core component of the E3-ubiquitin ligase, to promote

589

nuclear accumulation and transcriptional activity of Daf-16/FOXO. Studies in C. elegans

590

have also shown that Pek-1 acts synergistically with Atf-6 to maintain ER homeostasis as an

591

alternative mechanism to the Ire-1/Xbp-1 pathway. Crucially, Pek-1 and Atf-6 have both

592

been found to be able to compensate for the loss of ire-1 and xbp-1, which regulate the

593

same gene signatures as Daf-16/FOXO during ER stress response, suggesting a possible

594

central role of Daf-16/FOXO in maintaining ER homeostasis, which involves all three major

AC C

EP

TE D

M AN U

SC

RI PT

558

19

ACCEPTED MANUSCRIPT ER stress sensor-signalling pathways (Shen et al., 2005; Tatham et al., 2008; Zeng et al.,

596

2004). Daf-16 can also function independently of the three ER stress sensor-signalling

597

cascades by cooperating with Hsf-1 expression to activate chaprones such as Hsp70

598

which in turn promotes longevity and ER homeostasis through regulation of protein

599

folding(Hsu et al., 2003)

600

Regulation of PERK by dFOXO:

601

In a similar manner, a role for the fly protein kinase PERK in regulating dFOXO by

602

phosphorylation has also been described, consistent with its role in mammalian cells. PERK

603

constitutes another arm of the UPR, and inhibits protein biosynthesis under ER stress by

604

phosphorylation of the translation initiation factor eIF2α (Malhi and Kaufman, 2011).

605

However, it has been shown that PERK can also phosphorylate dFOXO at S243, promoting

606

its nuclear localization and activity, enhancing the ER stress response. Interestingly, this

607

contrasts with the effect of dFOXO phosphorylation by the Drosophila Akt, which results in

608

its sequestration in the cytoplasm (Zhang et al., 2013). In summary, in addition to studies in

609

mammals, studies in C. elegans and Drosophila models have provided clear evidence for

610

various crucial roles of the forkhead transcription factor FOXO in mediating the ER stress

611

response.

M AN U

SC

RI PT

595

612

Target cancer through the UPR signalling and its FOXO link

614

Accumulating evidence suggests that the ER-stress/UPR signalling network may be a

615

promising pharmacological target for targeting cancer cells and for overcoming drug

616

resistance, and several approaches are currently being pursued. The proposition that

617

disrupting cellular protein homeostasis by triggering overwhelming ER stress constitutes a

618

therapeutic option is supported by the success of proteasome inhibitors in some

619

hematological malignancies. The ubiquitin-proteasome pathway is the main biological

620

pathway for intracellular protein degradation, and inhibition of 26S proteasome can result in

621

an accumulation of defective and unfolded proteins within the ER. Proteasome inhibitors are

622

a novel class of cancer therapeutics designed to promote tumor cell specific cytotoxic effects

623

via inducing proteotoxic ER stress (Auner and Cenci, 2015; McConkey, 2017). Clinical trials

624

of these inhibitors, such as Bortezomib, have been conducted for a variety of malignancies,

625

resulting in drug regulatory agency approvals for the treatment of multiple myeloma and

626

mantle-cell lymphoma in patients (McConkey, 2017).

627

Targeting IRE1α-XBP1:

628

As the IRE1α-XBP1 signalling axis is central to cancer development, inhibition of IRE1α-

629

XBP1 pathway can provide a viable strategy for targeting cancer, and several specific

630

inhibitors are currently being explored in preclinical studies. For example, IRE1α-specific

AC C

EP

TE D

613

20

ACCEPTED MANUSCRIPT inhibitors have been proven to be able to trigger apoptosis of pancreatic cancer cells (Chien

632

et al., 2014). Another compound STF-083010, which can block the IRE1α endonuclease

633

function without influence on its kinase activity, also displays significant cytotoxic effects on

634

multiple myeloma cells although its selectivity not clear yet (Papandreou et al., 2010). 3-

635

Methoxy-5,6-dibromosalicylaldehyde and an IRE1α-binding small molecule 4u8C can inhibit

636

IRE1α-induced XBP1 splicing and RIDD, and sensitize the cells to ER stress and thus cell

637

death (Cross et al., 2012; Volkmann et al., 2011). The inhibitors toyocamycin and MKC-

638

3946, which target IRE1α-induced XBP1 splicing, have been demonstrated to be cytotoxic

639

for multiple myeloma cells (Mimura et al., 2012; Ri et al., 2012). In addition, inhibition of

640

IRE1α activity also enhances the efficacy of oncolytic viral therapy in vivo through the

641

induction of caspase-2-dependent cellular apoptosis (Mahoney et al., 2011). Moreover,

642

Imatinib-an anti-cancer tyrosine kinase inhibitor interrupts the e binding between ABL

643

tyrosine Kinase and IRE1α which leads to reverse type 1 diabetes in non-obese diabetic

644

(Morita et al., 2017).

645

Targeting PERK-ATF4:

646

In addition to IRE1α inhibitors, the realization that PERK over-activation has a significant

647

impact on development and progression of different types of cancers and the finding that

648

PERK knockdown prevents tumor growth inhibition in mice (Bi et al., 2005) have also led to

649

the generation of a number of small molecule inhibitors of PERK for cancer treatment (Pytel

650

et al., 2016). GSK2606414 is the first selective PERK inhibitor developed and has

651

demonstrated tumor growth inhibitory effects on human tumors in mouse xenograft models

652

(Chaturvedula et al., 2013). Hitherto, most PERK inhibitors have demonstrated therapeutic

653

potentials in restoring proteostasis, and in blocking tumor growth and angiogenesis in cell

654

culture and animal models of cancer (Axten, 2017).

655

Chaperones inhibitors and FOXO3:

656

The Hsp70 and Hsp90 ER-related chaperones plays a key role in the ER stress response

657

and are the first lines of defence in the cellular response against proteotoxic stress

658

(McConkey, 2017). Targeting these ER-related chaperones can potentially tip the UPR from

659

promoting cell survival towards cell termination.

660

Hsp90/GRP94 inhibitor FW-04-806 has been shown to display potent anti-tumor effects in

661

HER2-positive breast cancer (Huang et al., 2015). Accordingly, FW-04-806 can block the

662

PI3K/AKT pathway and induce FOXO3 to cause proliferative arrest and apoptosis in HER2+

663

breast cancer cells (Huang et al., 2015). Interestingly, FW-04-806 also combines

664

synergistically with the EGFR/HER2 tyrosine kinase inhibitor lapatinib (Huang et al., 2015),

665

which has previously been shown to target FOXO3 to mediate its cytostatic effects (Francis

666

et al., 2009; Krol et al., 2007). Moreover, in BRAF inhibitor-resistant melanoma cells, the

667

HSP90 inhibitor XL888 has also been found to prevent growth of tumor and lead to

AC C

EP

TE D

M AN U

SC

RI PT

631

21

Consistent with this concept, a novel

ACCEPTED MANUSCRIPT degradation of many survival signalling molecules, such as IGFR1, PDGFRβ and AKT

669

(Paraiso et al., 2012), which are upstream signalling repressors of FOXO3 (Myatt and Lam,

670

2007). In agreement, XL888 has been shown to mediate its cytotoxic function through

671

activating FOXO3 to induce the expression of the pro-apoptotic Bim and repress the

672

expression of the anti-apoptotic Mcl-1 Bcl-2-related genes (Paraiso et al., 2012).

673

Appropriately both Bim and Mcl-1 are downstream targets of FOXO3. In addition, 17-

674

Allylamino-17-demethoxygeldanamycin (17-AAG), a urinary bladder anti-cancer treatment,

675

induce apoptosis by disrupting the ability of Hsp90 to regulate its protein clients that regulate

676

cancer cells survival (Karkoulis et al., 2010). Since FOXO proteins play a crucial part in the

677

UPR, targeting the FOXO proteins can prove to be an attractive strategy for tackling cancer

678

and for overcoming drug resistance. FOXO activity is modulated by acetylation, which is in

679

turn regulated by Sir2-related NAD-dependent protein deacetylases, (SIRTs or Sirtuins)

680

(Olmos et al., 2011). Research from mammalian systems and nematodes has also shown

681

that SIRT-mediated FOXO deacetylation not only limits its cytotoxic activity but also confers

682

drug resistance by directing it towards genes that have a role in ER stress and

683

chemotherapy resistance (Khongkow et al., 2013; Olmos et al., 2011; Peck et al., 2010). A

684

number of SIRT inhibitors have now been developed and these compounds have

685

demonstrated to be able to enhance the cytotoxic function of FOXO3 in cancer drug

686

treatment (Khongkow et al., 2013; Olmos et al., 2011; Peck et al., 2010). Besides targeting

687

cancer independently, these UPR signalling disruption compounds may also prove useful to

688

reinforce the cytotoxic effects of chemotherapeutic drugs, particular in drug resistance

689

cancers. In addition to being anticancer therapeutic targets, these ER stress and FOXO-

690

regulated signalling and gene signatures can also be used as reliable diagnostic and

691

prognostic biomarkers and as indicators for drug responsiveness.

SC

M AN U

TE D

EP

692

RI PT

668

Acknowledgements

694

Glowi Alasiri is a recipient of a scholarship from the Saudi Arabian Cultural Bureau in

695

London (MSU434). Stefania Zona is a post-doctoral research associate supported by the

696

Medical Research Council (MRC) of UK (MR/N012097/1). Lavender Yuen-Nam Fan is a

697

PhD student and research assistant supported by Imperial College IC Trust. Holger W.

698

Auner acknowledges the support of the Imperial College London National Institute of Health

699

Research-Biomedical Research Centre (NIHR-BRC) and the Imperial College London

700

Cancer Research UK Centre. Eric W.-F. Lam's work is supported by MRC (MR/N012097/1),

701

CRUK (A12011), Breast Cancer Now (2012MayPR070; 2012NovPhD016), the Cancer

702

Research UK Imperial Centre, Imperial ECMC and NIHR Imperial BRC.

AC C

693

703

22

ACCEPTED MANUSCRIPT References

705 706 707 708 709 710 711 712 713 714 715 716 717 718 719 720 721 722 723 724 725 726 727 728 729 730 731 732 733 734 735 736 737 738 739 740 741 742 743 744 745 746 747 748 749 750 751 752 753 754

Anderson, M.J., Viars, C.S., Czekay, S., Cavenee, W.K. and Arden, K.C., 1998. Cloning and characterization of three human forkhead genes that comprise an FKHR‐like gene subfamily, Genomics. 47, 187‐199. Anelli, T. and Sitia, R., 2010. Physiology and pathology of proteostasis in the early secretory compartment, Semin Cell Dev Biol. 21, 520‐525. Araki, K. and Nagata, K., 2012. Protein Folding and Quality Control in the ER, Cold Spring Harbor Perspectives in Biology. 4, a015438‐a015438. Auner, H.W. and Cenci, S., 2015. Recent advances and future directions in targeting the secretory apparatus in multiple myeloma, Br J Haematol. 168, 14‐25. Axten, J.M., 2017. Protein kinase R(PKR)‐like endoplasmic reticulum kinase (PERK) inhibitors: a patent review (2010‐2015), Expert Opin Ther Pat. 27, 37‐48. B'Chir, W., Maurin, A.C., Carraro, V., Averous, J., Jousse, C., Muranishi, Y., Parry, L., Stepien, G., Fafournoux, P. and Bruhat, A., 2013. The eIF2 /ATF4 pathway is essential for stress‐induced autophagy gene expression, Nucleic Acids Research. 41, 7683‐7699. Baumann, J., Wong, J., Sun, Y. and Conklin, D.S., 2016. Palmitate‐induced ER stress increases trastuzumab sensitivity in HER2/neu‐positive breast cancer cells, BMC Cancer. 16, 551. Baumeister, P., Luo, S., Skarnes, W.C., Sui, G., Seto, E., Shi, Y. and Lee, A.S., 2005. Endoplasmic reticulum stress induction of the Grp78/BiP promoter: activating mechanisms mediated by YY1 and its interactive chromatin modifiers, Mol Cell Biol. 25, 4529‐4540. Bella, L., Zona, S., Nestal de Moraes, G. and Lam, E.W., 2014. FOXM1: A key oncofoetal transcription factor in health and disease, Semin Cancer Biol. 29, 32‐39. Bertolotti, A., Zhang, Y., Hendershot, L.M., Harding, H.P. and Ron, D., 2000. Dynamic interaction of BiP and ER stress transducers in the unfolded‐protein response, Nat Cell Biol. 2, 326‐332. Bi, M., Naczki, C., Koritzinsky, M., Fels, D., Blais, J., Hu, N., Harding, H., Novoa, I., Varia, M., Raleigh, J., Scheuner, D., Kaufman, R.J., Bell, J., Ron, D., Wouters, B.G. and Koumenis, C., 2005. ER stress‐regulated translation increases tolerance to extreme hypoxia and promotes tumor growth, EMBO J. 24, 3470‐3481. Blais, J.D., Addison, C.L., Edge, R., Falls, T., Zhao, H., Wary, K., Koumenis, C., Harding, H.P., Ron, D., Holcik, M. and Bell, J.C., 2006. Perk‐dependent translational regulation promotes tumor cell adaptation and angiogenesis in response to hypoxic stress, Mol Cell Biol. 26, 9517‐9532. Bobrovnikova‐Marjon, E., Grigoriadou, C., Pytel, D., Zhang, F., Ye, J., Koumenis, C., Cavener, D. and Diehl, J.A., 2010. PERK promotes cancer cell proliferation and tumor growth by limiting oxidative DNA damage, Oncogene. 29, 3881‐3895. Bobrovnikova‐Marjon, E., Pytel, D., Riese, M.J., Vaites, L.P., Singh, N., Koretzky, G.A., Witze, E.S. and Diehl, J.A., 2012. PERK Utilizes Intrinsic Lipid Kinase Activity To Generate Phosphatidic Acid, Mediate Akt Activation, and Promote Adipocyte Differentiation, Molecular and Cellular Biology. 32, 2268‐2278. Braakman, I. and Hebert, D.N., 2013. Protein folding in the endoplasmic reticulum, Cold Spring Harb Perspect Biol. 5, a013201. Bu, Y. and Diehl, J.A., 2016. PERK Integrates Oncogenic Signaling and Cell Survival During Cancer Development, J Cell Physiol. Calfon, M., Zeng, H., Urano, F., Till, J.H., Hubbard, S.R., Harding, H.P., Clark, S.G. and Ron, D., 2002. IRE1 couples endoplasmic reticulum load to secretory capacity by processing the XBP‐1 mRNA, Nature. 415, 92‐96. Calnan, D.R. and Brunet, A., 2008. The FoxO code, Oncogene. 27, 2276‐2288. Carrasco, D.R., Sukhdeo, K., Protopopova, M., Sinha, R., Enos, M., Carrasco, Daniel E., Zheng, M., Mani, M., Henderson, J., Pinkus, G.S., Munshi, N., Horner, J., Ivanova, E.V., Protopopov, A., Anderson, K.C., Tonon, G. and DePinho, R.A., 2007. The Differentiation and Stress Response Factor XBP‐1 Drives Multiple Myeloma Pathogenesis, Cancer Cell. 11, 349‐360.

AC C

EP

TE D

M AN U

SC

RI PT

704

23

ACCEPTED MANUSCRIPT

EP

TE D

M AN U

SC

RI PT

Caruso, M.E., Jenna, S., Bouchecareilh, M., Baillie, D.L., Boismenu, D., Halawani, D., Latterich, M. and Chevet, E., 2008. GTPase‐mediated regulation of the unfolded protein response in Caenorhabditis elegans is dependent on the AAA+ ATPase CDC‐48, Mol Cell Biol. 28, 4261‐ 4274. Chambers, J.E., Petrova, K., Tomba, G., Vendruscolo, M. and Ron, D., 2012. ADP ribosylation adapts an ER chaperone response to short‐term fluctuations in unfolded protein load, J Cell Biol. 198, 371‐385. Chang, Y.‐J., Huang, Y.‐P., Li, Z.‐L. and Chen, C.‐H., 2012. GRP78 Knockdown Enhances Apoptosis via the Down‐Regulation of Oxidative Stress and Akt Pathway after Epirubicin Treatment in Colon Cancer DLD‐1 Cells, PLoS ONE. 7, e35123. Chaturvedula, P.V., Mercer, S.E., Pin, S.S., Thalody, G., Xu, C., Conway, C.M., Keavy, D., Signor, L., Cantor, G.H., Mathias, N., Moench, P., Denton, R., Macci, R., Schartman, R., Whiterock, V., Davis, C., Macor, J.E. and Dubowchik, G.M., 2013. Discovery of (R)‐N‐(3‐(7‐methyl‐1H‐ indazol‐5‐yl)‐1‐(4‐(1‐methylpiperidin‐4‐yl)‐1‐oxopropan‐2‐y l)‐4‐(2‐oxo‐1,2‐dihydroquinolin‐ 3‐yl)piperidine‐1‐carboxamide (BMS‐742413): a potent human CGRP antagonist with superior safety profile for the treatment of migraine through intranasal delivery, Bioorg Med Chem Lett. 23, 3157‐3161. Chien, W., Ding, L.‐W., Sun, Q.‐Y., Torres‐Fernandez, L.A., Tan, S.Z., Xiao, J., Lim, S.L., Garg, M., Lee, K.L., Kitajima, S., Takao, S., Leong, W.Z., Sun, H., Tokatly, I., Poellinger, L., Gery, S. and Koeffler, P.H., 2014. Selective inhibition of unfolded protein response induces apoptosis in pancreatic cancer cells, Oncotarget. 5, 4881‐4894. Chillaron, J. and Haas, I.G., 2000. Dissociation from BiP and retrotranslocation of unassembled immunoglobulin light chains are tightly coupled to proteasome activity, Mol Biol Cell. 11, 217‐226. Chitnis, N.S., Pytel, D., Bobrovnikova‐Marjon, E., Pant, D., Zheng, H., Maas, N.L., Frederick, B., Kushner, J.A., Chodosh, L.A., Koumenis, C., Fuchs, S.Y. and Diehl, J.A., 2012. miR‐211 is a prosurvival microRNA that regulates chop expression in a PERK‐dependent manner, Mol Cell. 48, 353‐364. Clarke, R., Cook, K.L., Hu, R., Facey, C.O.B., Tavassoly, I., Schwartz, J.L., Baumann, W.T., Tyson, J.J., Xuan, J., Wang, Y., Warri, A. and Shajahan, A.N., 2012. Endoplasmic Reticulum Stress, the Unfolded Protein Response, Autophagy, and the Integrated Regulation of Breast Cancer Cell Fate, Cancer Research. Colvin, T.A., Gabai, V.L., Gong, J., Calderwood, S.K., Li, H., Gummuluru, S., Matchuk, O.N., Smirnova, S.G., Orlova, N.V., Zamulaeva, I.A., Garcia‐Marcos, M., Li, X., Young, Z.T., Rauch, J.N., Gestwicki, J.E., Takayama, S. and Sherman, M.Y., 2014. Hsp70‐Bag3 interactions regulate cancer‐related signaling networks, Cancer Res. 74, 4731‐4740. Consolaro, F., Ghaem‐Maghami, S., Bortolozzi, R., Zona, S., Khongkow, M., Basso, G., Viola, G. and Lam, E.W., 2015. FOXO3a and Posttranslational Modifications Mediate Glucocorticoid Sensitivity in B‐ALL, Mol Cancer Res. 13, 1578‐1590. Corazzari, M., Rapino, F., Ciccosanti, F., Giglio, P., Antonioli, M., Conti, B., Fimia, G.M., Lovat, P.E. and Piacentini, M., 2015. Oncogenic BRAF induces chronic ER stress condition resulting in increased basal autophagy and apoptotic resistance of cutaneous melanoma, Cell Death Differ. 22, 946‐958. Cross, B.C.S., Bond, P.J., Sadowski, P.G., Jha, B.K., Zak, J., Goodman, J.M., Silverman, R.H., Neubert, T.A., Baxendale, I.R., Ron, D. and Harding, H.P., 2012. The molecular basis for selective inhibition of unconventional mRNA splicing by an IRE1‐binding small molecule, Proceedings of the National Academy of Sciences. 109, E869‐E878. Dang, H., Klokk, T.I., Schaheen, B., McLaughlin, B.M., Thomas, A.J., Durns, T.A., Bitler, B.G., Sandvig, K. and Fares, H., 2011. Derlin‐dependent retrograde transport from endosomes to the Golgi apparatus, Traffic. 12, 1417‐1431.

AC C

755 756 757 758 759 760 761 762 763 764 765 766 767 768 769 770 771 772 773 774 775 776 777 778 779 780 781 782 783 784 785 786 787 788 789 790 791 792 793 794 795 796 797 798 799 800 801 802 803 804

24

ACCEPTED MANUSCRIPT

EP

TE D

M AN U

SC

RI PT

Darom, A., Bening‐Abu‐Shach, U. and Broday, L., 2010. RNF‐121 is an endoplasmic reticulum‐ membrane E3 ubiquitin ligase involved in the regulation of beta‐integrin, Mol Biol Cell. 21, 1788‐1798. David, V., Hochstenbach, F., Rajagopalan, S. and Brenner, M.B., 1993. Interaction with newly synthesized and retained proteins in the endoplasmic reticulum suggests a chaperone function for human integral membrane protein IP90 (calnexin), J Biol Chem. 268, 9585‐9592. Deshaies, R.J., 2014. Proteotoxic crisis, the ubiquitin‐proteasome system, and cancer therapy, BMC Biol. 12, 94. Donovan, M.R. and Marr, M.T., 2nd, 2016. dFOXO Activates Large and Small Heat Shock Protein Genes in Response to Oxidative Stress to Maintain Proteostasis in Drosophila, J Biol Chem. 291, 19042‐19050. Dufey, E., Sepulveda, D., Rojas‐Rivera, D. and Hetz, C., 2014. Cellular mechanisms of endoplasmic reticulum stress signaling in health and disease. 1. An overview, Am J Physiol Cell Physiol. 307, C582‐594. Feng, H., Zhong, W., Punkosdy, G., Gu, S., Zhou, L., Seabolt, E.K. and Kipreos, E.T., 1999. CUL‐2 is required for the G1‐to‐S‐phase transition and mitotic chromosome condensation in Caenorhabditis elegans, Nat Cell Biol. 1, 486‐492. Fernandes, M., Xiao, H. and Lis, J.T., 1995. Binding of heat shock factor to and transcriptional activation of heat shock genes in Drosophila, Nucleic Acids Res. 23, 4799‐4804. Francis, R.E., Myatt, S.S., Krol, J., Hartman, J., Peck, B., McGovern, U.B., Wang, J., Guest, S.K., Filipovic, A., Gojis, O., Palmieri, C., Peston, D., Shousha, S., Yu, Q., Sicinski, P., Coombes, R.C. and Lam, E.W., 2009. FoxM1 is a downstream target and marker of HER2 overexpression in breast cancer, Int J Oncol. 35, 57‐68. Frand, A.R. and Kaiser, C.A., 1999. Ero1p oxidizes protein disulfide isomerase in a pathway for disulfide bond formation in the endoplasmic reticulum, Mol Cell. 4, 469‐477. Fu, Z. and Tindall, D.J., 2008. FOXOs, cancer and regulation of apoptosis, Oncogene. 27, 2312‐2319. Fujimoto, T., Onda, M., Nagai, H., Nagahata, T., Ogawa, K. and Emi, M., 2003. Upregulation and overexpression of human x‐box binding protein 1 (hXBP‐1) gene in primary breast cancers, Breast Cancer. 10, 301‐306. Furuyama, T., Nakazawa, T., Nakano, I. and Mori, N., 2000. Identification of the differential distribution patterns of mRNAs and consensus binding sequences for mouse DAF‐16 homologues, Biochem J. 349, 629‐634. Gardner, B.M. and Walter, P., 2011. Unfolded Proteins are Ire1‐Activating Ligands that Directly Induce the Unfolded Protein Response, Science (New York, N.Y.). 333, 1891‐1894. Ghazi, A., Henis‐Korenblit, S. and Kenyon, C., 2007. Regulation of Caenorhabditis elegans lifespan by a proteasomal E3 ligase complex, Proc Natl Acad Sci U S A. 104, 5947‐5952. Ghosh, A.P., Klocke, B.J., Ballestas, M.E. and Roth, K.A., 2012. CHOP potentially co‐operates with FOXO3a in neuronal cells to regulate PUMA and BIM expression in response to ER stress, PLoS One. 7, e39586. Ghosh, H.S., Reizis, B. and Robbins, P.D., 2011. SIRT1 associates with eIF2‐alpha and regulates the cellular stress response, Sci Rep. 1, 150. Ghosh, R., Lipson, K.L., Sargent, K.E., Mercurio, A.M., Hunt, J.S., Ron, D. and Urano, F., 2010. Transcriptional Regulation of VEGF‐A by the Unfolded Protein Response Pathway, PLoS ONE. 5, e9575. Glab, J.A., Doerflinger, M., Nedeva, C., Jose, I., Mbogo, G.W., Paton, J.C., Paton, A.W., Kueh, A.J., Herold, M.J., Huang, D.C., Segal, D., Brumatti, G. and Puthalakath, H., 2017. DR5 and caspase‐8 are dispensable in ER stress‐induced apoptosis, Cell Death Differ. 24, 944‐950. Gomez‐Puerto, M.C., Verhagen, L.P., Braat, A.K., Lam, E.W., Coffer, P.J. and Lorenowicz, M.J., 2016. Activation of autophagy by FOXO3 regulates redox homeostasis during osteogenic differentiation, Autophagy. 12, 1804‐1816.

AC C

805 806 807 808 809 810 811 812 813 814 815 816 817 818 819 820 821 822 823 824 825 826 827 828 829 830 831 832 833 834 835 836 837 838 839 840 841 842 843 844 845 846 847 848 849 850 851 852 853 854

25

ACCEPTED MANUSCRIPT

EP

TE D

M AN U

SC

RI PT

Greer, E.L. and Brunet, A., 2005. FOXO transcription factors at the interface between longevity and tumor suppression, Oncogene. 24, 7410‐7425. Greer, E.L., Oskoui, P.R., Banko, M.R., Maniar, J.M., Gygi, M.P., Gygi, S.P. and Brunet, A., 2007. The energy sensor AMP‐activated protein kinase directly regulates the mammalian FOXO3 transcription factor, J Biol Chem. 282, 30107‐30119. Hagiwara, M., Maegawa, K., Suzuki, M., Ushioda, R., Araki, K., Matsumoto, Y., Hoseki, J., Nagata, K. and Inaba, K., 2011. Structural basis of an ERAD pathway mediated by the ER‐resident protein disulfide reductase ERdj5, Mol Cell. 41, 432‐444. Han, J., Back, S.H., Hur, J., Lin, Y.‐H., Gildersleeve, R., Shan, J., Yuan, C.L., Krokowski, D., Wang, S., Hatzoglou, M., Kilberg, M.S., Sartor, M.A. and Kaufman, R.J., 2013. ER‐stress‐induced transcriptional regulation increases protein synthesis leading to cell death, Nature Cell Biology. 15, 481‐490. Han, J., Back, S.H., Hur, J., Lin, Y.H., Gildersleeve, R., Shan, J., Yuan, C.L., Krokowski, D., Wang, S., Hatzoglou, M., Kilberg, M.S., Sartor, M.A. and Kaufman, R.J., 2013. ER‐stress‐induced transcriptional regulation increases protein synthesis leading to cell death, Nat Cell Biol. 15, 481‐490. Harding, H.P., Novoa, I., Zhang, Y., Zeng, H., Wek, R., Schapira, M. and Ron, D., 2000. Regulated translation initiation controls stress‐induced gene expression in mammalian cells, Mol Cell. 6, 1099‐1108. Harding, H.P., Zhang, Y., Zeng, H., Novoa, I., Lu, P.D., Calfon, M., Sadri, N., Yun, C., Popko, B., Paules, R., Stojdl, D.F., Bell, J.C., Hettmann, T., Leiden, J.M. and Ron, D., 2003. An integrated stress response regulates amino acid metabolism and resistance to oxidative stress, Mol Cell. 11, 619‐633. Hart, L.S., Cunningham, J.T., Datta, T., Dey, S., Tameire, F., Lehman, S.L., Qiu, B., Zhang, H., Cerniglia, G., Bi, M., Li, Y., Gao, Y., Liu, H., Li, C., Maity, A., Thomas‐Tikhonenko, A., Perl, A.E., Koong, A., Fuchs, S.Y., Diehl, J.A., Mills, I.G., Ruggero, D. and Koumenis, C., 2012. ER stress‐mediated autophagy promotes Myc‐dependent transformation and tumor growth, J Clin Invest. 122, 4621‐4634. Hart, L.S., Cunningham, J.T., Datta, T., Dey, S., Tameire, F., Lehman, S.L., Qiu, B., Zhang, H., Cerniglia, G., Bi, M., Li, Y., Gao, Y., Liu, H., Li, C., Maity, A., Thomas‐Tikhonenko, A., Perl, A.E., Koong, A., Fuchs, S.Y., Diehl, J.A., Mills, I.G., Ruggero, D. and Koumenis, C., 2012. ER stress–mediated autophagy promotes Myc‐dependent transformation and tumor growth, Journal of Clinical Investigation. 122, 4621‐4634. Hartl, F.U. and Hayer‐Hartl, M., 2002. Molecular chaperones in the cytosol: from nascent chain to folded protein, Science. 295, 1852‐1858. Henis‐Korenblit, S., Zhang, P., Hansen, M., McCormick, M., Lee, S.J., Cary, M. and Kenyon, C., 2010. Insulin/IGF‐1 signaling mutants reprogram ER stress response regulators to promote longevity, Proc Natl Acad Sci U S A. 107, 9730‐9735. Hetz, C., 2012. The unfolded protein response: controlling cell fate decisions under ER stress and beyond, Nat Rev Mol Cell Biol. 13, 89‐102. Hetz, C., Bernasconi, P., Fisher, J., Lee, A.H., Bassik, M.C., Antonsson, B., Brandt, G.S., Iwakoshi, N.N., Schinzel, A., Glimcher, L.H. and Korsmeyer, S.J., 2006. Proapoptotic BAX and BAK modulate the unfolded protein response by a direct interaction with IRE1alpha, Science. 312, 572‐576. Ho, K.K., McGuire, V.A., Koo, C.Y., Muir, K.W., de Olano, N., Maifoshie, E., Kelly, D.J., McGovern, U.B., Monteiro, L.J., Gomes, A.R., Nebreda, A.R., Campbell, D.G., Arthur, J.S. and Lam, E.W., 2012. Phosphorylation of FOXO3a on Ser‐7 by p38 promotes its nuclear localization in response to doxorubicin, J Biol Chem. 287, 1545‐1555. Hollien, J., Lin, J.H., Li, H., Stevens, N., Walter, P. and Weissman, J.S., 2009. Regulated Ire1‐ dependent decay of messenger RNAs in mammalian cells, J Cell Biol. 186, 323‐331. Hou, X., Liu, Y., Liu, H., Chen, X., Liu, M., Che, H., Guo, F., Wang, C., Zhang, D., Wu, J., Chen, X., Shen, C., Li, C., Peng, F., Bi, Y., Yang, Z., Yang, G., Ai, J., Gao, X. and Zhao, S., 2015. PERK silence

AC C

855 856 857 858 859 860 861 862 863 864 865 866 867 868 869 870 871 872 873 874 875 876 877 878 879 880 881 882 883 884 885 886 887 888 889 890 891 892 893 894 895 896 897 898 899 900 901 902 903 904 905

26

ACCEPTED MANUSCRIPT

EP

TE D

M AN U

SC

RI PT

inhibits glioma cell growth under low glucose stress by blockage of p‐AKT and subsequent HK2's mitochondria translocation, Sci Rep. 5, 9065. Høyer‐Hansen, M. and Jäättelä, M., 2007. Connecting endoplasmic reticulum stress to autophagy by unfolded protein response and calcium, Cell Death Differ. 14, 1576‐1582. Hsin, I.L., Hsiao, Y.C., Wu, M.F., Jan, M.S., Tang, S.C., Lin, Y.W., Hsu, C.P. and Ko, J.L., 2012. Lipocalin 2, a new GADD153 target gene, as an apoptosis inducer of endoplasmic reticulum stress in lung cancer cells, Toxicol Appl Pharmacol. 263, 330‐337. Hsu, A.L., Murphy, C.T. and Kenyon, C., 2003. Regulation of aging and age‐related disease by DAF‐16 and heat‐shock factor, Science. 300, 1142‐1145. Huang, W., Wu, Q.D., Zhang, M., Kong, Y.L., Cao, P.R., Zheng, W., Xu, J.H. and Ye, M., 2015. Novel Hsp90 inhibitor FW‐04‐806 displays potent antitumor effects in HER2‐positive breast cancer cells as a single agent or in combination with lapatinib, Cancer Lett. 356, 862‐871. Huggins, C.J., Mayekar, M.K., Martin, N., Saylor, K.L., Gonit, M., Jailwala, P., Kasoji, M., Haines, D.C., Quinones, O.A. and Johnson, P.F., 2015. C/EBPgamma Is a Critical Regulator of Cellular Stress Response Networks through Heterodimerization with ATF4, Mol Cell Biol. 36, 693‐713. Janiesch, P.C., Kim, J., Mouysset, J., Barikbin, R., Lochmuller, H., Cassata, G., Krause, S. and Hoppe, T., 2007. The ubiquitin‐selective chaperone CDC‐48/p97 links myosin assembly to human myopathy, Nat Cell Biol. 9, 379‐390. Jansen, G., Maattanen, P., Denisov, A.Y., Scarffe, L., Schade, B., Balghi, H., Dejgaard, K., Chen, L.Y., Muller, W.J., Gehring, K. and Thomas, D.Y., 2012. An interaction map of endoplasmic reticulum chaperones and foldases, Mol Cell Proteomics. 11, 710‐723. Kalvakolanu, D.V. and Gade, P., 2012. IFNG and autophagy, Autophagy. 8, 1673‐1674. Kapulkin, W.J., Hiester, B.G. and Link, C.D., 2005. Compensatory regulation among ER chaperones in C. elegans, FEBS Lett. 579, 3063‐3068. Karkoulis, P.K., Stravopodis, D.J., Margaritis, L.H. and Voutsinas, G.E., 2010. 17‐Allylamino‐17‐ demethoxygeldanamycin induces downregulation of critical Hsp90 protein clients and results in cell cycle arrest and apoptosis of human urinary bladder cancer cells, BMC Cancer. 10, 481. Karunarathna, U., Kongsema, M., Zona, S., Gong, C., Cabrera, E., Gomes, A.R., Man, E.P., Khongkow, P., Tsang, J.W., Khoo, U.S., Medema, R.H., Freire, R. and Lam, E.W., 2016. OTUB1 inhibits the ubiquitination and degradation of FOXM1 in breast cancer and epirubicin resistance, Oncogene. 35, 1433‐1444. Kaufman, R.J., 1999. Stress signaling from the lumen of the endoplasmic reticulum: coordination of gene transcriptional and translational controls, Genes Dev. 13, 1211‐1233. Kaushik, S. and Cuervo, A.M., 2015. Proteostasis and aging, Nat Med. 21, 1406‐1415. Khongkow, M., Olmos, Y., Gong, C., Gomes, A.R., Monteiro, L.J., Yague, E., Cavaco, T.B., Khongkow, P., Man, E.P., Laohasinnarong, S., Koo, C.Y., Harada‐Shoji, N., Tsang, J.W., Coombes, R.C., Schwer, B., Khoo, U.S. and Lam, E.W., 2013. SIRT6 modulates paclitaxel and epirubicin resistance and survival in breast cancer, Carcinogenesis. 34, 1476‐1486. Khongkow, P., Gomes, A.R., Gong, C., Man, E.P., Tsang, J.W., Zhao, F., Monteiro, L.J., Coombes, R.C., Medema, R.H., Khoo, U.S. and Lam, E.W., 2015. Paclitaxel targets FOXM1 to regulate KIF20A in mitotic catastrophe and breast cancer paclitaxel resistance Oncogene. 34, DOI: 10.1038/onc.2015.1152. Khongkow, P., Karunarathna, U., Khongkow, M., Gong, C., Gomes, A.R., Yague, E., Monteiro, L.J., Kongsema, M., Zona, S., Man, E.P., Tsang, J.W., Coombes, R.C., Wu, K.J., Khoo, U.S., Medema, R.H., Freire, R. and Lam, E.W., 2014. FOXM1 targets NBS1 to regulate DNA damage‐induced senescence and epirubicin resistance, Oncogene. 33, 4144‐4155. Koo, C.‐Y., Muir, K.W. and Lam, E.W.F., 2012. FOXM1: From cancer initiation to progression and treatment, Biochimica et Biophysica Acta (BBA) ‐ Gene Regulatory Mechanisms. 1819, 28‐37. Koo, C.Y., Muir, K.W. and Lam, E.W., 2012. FOXM1: From cancer initiation to progression and treatment, Biochim Biophys Acta. 1819, 28‐37.

AC C

906 907 908 909 910 911 912 913 914 915 916 917 918 919 920 921 922 923 924 925 926 927 928 929 930 931 932 933 934 935 936 937 938 939 940 941 942 943 944 945 946 947 948 949 950 951 952 953 954 955 956

27

ACCEPTED MANUSCRIPT

EP

TE D

M AN U

SC

RI PT

Kozutsumi, Y., Segal, M., Normington, K., Gething, M.J. and Sambrook, J., 1988. The presence of malfolded proteins in the endoplasmic reticulum signals the induction of glucose‐regulated proteins, Nature. 332, 462‐464. Krol, J., Francis, R.E., Albergaria, A., Sunters, A., Polychronis, A., Coombes, R.C. and Lam, E.W., 2007. The transcription factor FOXO3a is a crucial cellular target of gefitinib (Iressa) in breast cancer cells, Mol Cancer Ther. 6, 3169‐3179. Kwok, J.M., Peck, B., Monteiro, L.J., Schwenen, H.D., Millour, J., Coombes, R.C., Myatt, S.S. and Lam, E.W., 2010. FOXM1 confers acquired cisplatin resistance in breast cancer cells, Mol Cancer Res. 8, 24‐34. Lam, E.W., Brosens, J.J., Gomes, A.R. and Koo, C.Y., 2013. Forkhead box proteins: tuning forks for transcriptional harmony, Nat Rev Cancer. 13, 482‐495. Lee, A.H., Iwakoshi, N.N. and Glimcher, L.H., 2003. XBP‐1 regulates a subset of endoplasmic reticulum resident chaperone genes in the unfolded protein response, Mol Cell Biol. 23, 7448‐7459. Lee, A.S., 2005. The ER chaperone and signaling regulator GRP78/BiP as a monitor of endoplasmic reticulum stress, Methods. 35, 373‐381. Lee, D., Jeong, D.E., Son, H.G., Yamaoka, Y., Kim, H., Seo, K., Khan, A.A., Roh, T.Y., Moon, D.W., Lee, Y. and Lee, S.J., 2015. SREBP and MDT‐15 protect C. elegans from glucose‐induced accelerated aging by preventing accumulation of saturated fat, Genes Dev. 29, 2490‐2503. Lee, K., Tirasophon, W., Shen, X., Michalak, M., Prywes, R., Okada, T., Yoshida, H., Mori, K. and Kaufman, R.J., 2002. IRE1‐mediated unconventional mRNA splicing and S2P‐mediated ATF6 cleavage merge to regulate XBP1 in signaling the unfolded protein response, Genes Dev. 16, 452‐466. Li, J., Lee, B. and Lee, A.S., 2006. Endoplasmic reticulum stress‐induced apoptosis: multiple pathways and activation of p53‐up‐regulated modulator of apoptosis (PUMA) and NOXA by p53, J Biol Chem. 281, 7260‐7270. Li, Z., Bridges, B., Olson, J. and Weinman, S.A., 2016. The interaction between acetylation and serine‐ 574 phosphorylation regulates the apoptotic function of FOXO3, Oncogene. Lin, K., Hsin, H., Libina, N. and Kenyon, C., 2001. Regulation of the Caenorhabditis elegans longevity protein DAF‐16 by insulin/IGF‐1 and germline signaling, Nat Genet. 28, 139‐145. Lin, L., Hron, J.D. and Peng, S.L., 2004. Regulation of NF‐kappaB, Th activation, and autoinflammation by the forkhead transcription factor Foxo3a, Immunity. 21, 203‐213. Liu, G., Rogers, J., Murphy, C.T. and Rongo, C., 2011. EGF signalling activates the ubiquitin proteasome system to modulate C. elegans lifespan, EMBO J. 30, 2990‐3003. Liu, J., Vasudevan, S. and Kipreos, E.T., 2004. CUL‐2 and ZYG‐11 promote meiotic anaphase II and the proper placement of the anterior‐posterior axis in C. elegans, Development. 131, 3513‐3525. Liu, L., Qi, X., Chen, Z., Shaw, L., Cai, J., Smith, L.H., Grant, M.B. and Boulton, M.E., 2013. Targeting the IRE1α/XBP1 and ATF6 Arms of the Unfolded Protein Response Enhances VEGF Blockade to Prevent Retinal and Choroidal Neovascularization, The American Journal of Pathology. 182, 1412‐1424. Luo, X., Yao, J., Nie, P., Yang, Z., Feng, H., Chen, P., Shi, X. and Zou, Z., 2016. FOXM1 promotes invasion and migration of colorectal cancer cells partially dependent on HSPA5 transactivation, Oncotarget. 7, 26480‐26495. Mahoney, Douglas J., Lefebvre, C., Allan, K., Brun, J., Sanaei, Cina A., Baird, S., Pearce, N., Grönberg, S., Wilson, B., Prakesh, M., Aman, A., Isaac, M., Mamai, A., Uehling, D., Al‐Awar, R., Falls, T., Alain, T. and Stojdl, David F., 2011. Virus‐Tumor Interactome Screen Reveals ER Stress Response Can Reprogram Resistant Cancers for Oncolytic Virus‐Triggered Caspase‐2 Cell Death, Cancer Cell. 20, 443‐456. Malhi, H. and Kaufman, R.J., 2011. Endoplasmic reticulum stress in liver disease, J Hepatol. 54, 795‐ 809.

AC C

957 958 959 960 961 962 963 964 965 966 967 968 969 970 971 972 973 974 975 976 977 978 979 980 981 982 983 984 985 986 987 988 989 990 991 992 993 994 995 996 997 998 999 1000 1001 1002 1003 1004 1005 1006

28

ACCEPTED MANUSCRIPT

EP

TE D

M AN U

SC

RI PT

Maurel, M., Chevet, E., Tavernier, J. and Gerlo, S., 2014. Getting RIDD of RNA: IRE1 in cell fate regulation, Trends in Biochemical Sciences. 39, 245‐254. McConkey, D.J., 2017. The integrated stress response and proteotoxicity in cancer therapy, Biochem Biophys Res Commun. 482, 450‐453. Mehnert, M., Sommer, T. and Jarosch, E., 2014. Der1 promotes movement of misfolded proteins through the endoplasmic reticulum membrane, Nat Cell Biol. 16, 77‐86. Meyer, H., Bug, M. and Bremer, S., 2012. Emerging functions of the VCP/p97 AAA‐ATPase in the ubiquitin system, Nat Cell Biol. 14, 117‐123. Miao, Y.R., Eckhardt, B.L., Cao, Y., Pasqualini, R., Argani, P., Arap, W., Ramsay, R.G. and Anderson, R.L., 2013. Inhibition of established micrometastases by targeted drug delivery via cell surface‐associated GRP78, Clin Cancer Res. 19, 2107‐2116. Milani, M., Rzymski, T., Mellor, H.R., Pike, L., Bottini, A., Generali, D. and Harris, A.L., 2009. The Role of ATF4 Stabilization and Autophagy in Resistance of Breast Cancer Cells Treated with Bortezomib, Cancer Research. 69, 4415‐4423. Mimura, N., Fulciniti, M., Gorgun, G., Tai, Y.T., Cirstea, D., Santo, L., Hu, Y., Fabre, C., Minami, J., Ohguchi, H., Kiziltepe, T., Ikeda, H., Kawano, Y., French, M., Blumenthal, M., Tam, V., Kertesz, N.L., Malyankar, U.M., Hokenson, M., Pham, T., Zeng, Q., Patterson, J.B., Richardson, P.G., Munshi, N.C. and Anderson, K.C., 2012. Blockade of XBP1 splicing by inhibition of IRE1 is a promising therapeutic option in multiple myeloma, Blood. 119, 5772‐5781. Molinari, M. and Helenius, A., 1999. Glycoproteins form mixed disulphides with oxidoreductases during folding in living cells, Nature. 402, 90‐93. Monteiro, L.J., Khongkow, P., Kongsema, M., Morris, J.R., Man, C., Weekes, D., Koo, C.Y., Gomes, A.R., Pinto, P.H., Varghese, V., Kenny, L.M., Charles Coombes, R., Freire, R., Medema, R.H. and Lam, E.W., 2013. The Forkhead Box M1 protein regulates BRIP1 expression and DNA damage repair in epirubicin treatment, Oncogene. 32, 4634‐4645. Morita, S., Villalta, S.A., Feldman, H.C., Register, A.C., Rosenthal, W., Hoffmann‐Petersen, I.T., Mehdizadeh, M., Ghosh, R., Wang, L., Colon‐Negron, K., Meza‐Acevedo, R., Backes, B.J., Maly, D.J., Bluestone, J.A. and Papa, F.R., 2017. Targeting ABL‐IRE1alpha Signaling Spares ER‐ Stressed Pancreatic beta Cells to Reverse Autoimmune Diabetes, Cell Metab. 25, 883‐ 897.e888. Morris, J.A., Dorner, A.J., Edwards, C.A., Hendershot, L.M. and Kaufman, R.J., 1997. Immunoglobulin Binding Protein (BiP) Function Is Required to Protect Cells from Endoplasmic Reticulum Stress but Is Not Required for the Secretion of Selective Proteins, Journal of Biological Chemistry. 272, 4327‐4334. Munoz‐Lobato, F., Rodriguez‐Palero, M.J., Naranjo‐Galindo, F.J., Shephard, F., Gaffney, C.J., Szewczyk, N.J., Hamamichi, S., Caldwell, K.A., Caldwell, G.A., Link, C.D. and Miranda‐Vizuete, A., 2014. Protective role of DNJ‐27/ERdj5 in Caenorhabditis elegans models of human neurodegenerative diseases, Antioxid Redox Signal. 20, 217‐235. Murphy, C.T., McCarroll, S.A., Bargmann, C.I., Fraser, A., Kamath, R.S., Ahringer, J., Li, H. and Kenyon, C., 2003. Genes that act downstream of DAF‐16 to influence the lifespan of Caenorhabditis elegans, Nature. 424, 277‐283. Myatt, S.S., Kongsema, M., Man, C.W., Kelly, D.J., Gomes, A.R., Khongkow, P., Karunarathna, U., Zona, S., Langer, J.K., Dunsby, C.W., Coombes, R.C., French, P.M., Brosens, J.J. and Lam, E.W., 2014. SUMOylation inhibits FOXM1 activity and delays mitotic transition, Oncogene. 33, 4316‐4329. Myatt, S.S. and Lam, E.W., 2007. The emerging roles of forkhead box (Fox) proteins in cancer, Nat Rev Cancer. 7, 847‐859. Myatt, S.S. and Lam, E.W.F., 2007. The emerging roles of forkhead box (Fox) proteins in cancer, Nature Reviews Cancer. 7, 847‐859. Nakanishi, K., Sudo, T. and Morishima, N., 2005. Endoplasmic reticulum stress signaling transmitted by ATF6 mediates apoptosis during muscle development, J Cell Biol. 169, 555‐560.

AC C

1007 1008 1009 1010 1011 1012 1013 1014 1015 1016 1017 1018 1019 1020 1021 1022 1023 1024 1025 1026 1027 1028 1029 1030 1031 1032 1033 1034 1035 1036 1037 1038 1039 1040 1041 1042 1043 1044 1045 1046 1047 1048 1049 1050 1051 1052 1053 1054 1055 1056 1057

29

ACCEPTED MANUSCRIPT

EP

TE D

M AN U

SC

RI PT

Nakatsukasa, K. and Brodsky, J.L., 2008. The recognition and retrotranslocation of misfolded proteins from the endoplasmic reticulum, Traffic. 9, 861‐870. Nayak, S., Santiago, F.E., Jin, H., Lin, D., Schedl, T. and Kipreos, E.T., 2002. The Caenorhabditis elegans Skp1‐related gene family: diverse functions in cell proliferation, morphogenesis, and meiosis, Curr Biol. 12, 277‐287. Nishikori, S., Yamanaka, K., Sakurai, T., Esaki, M. and Ogura, T., 2008. p97 Homologs from Caenorhabditis elegans, CDC‐48.1 and CDC‐48.2, suppress the aggregate formation of huntingtin exon1 containing expanded polyQ repeat, Genes Cells. 13, 827‐838. Nishitoh, H., Matsuzawa, A., Tobiume, K., Saegusa, K., Takeda, K., Inoue, K., Hori, S., Kakizuka, A. and Ichijo, H., 2002. ASK1 is essential for endoplasmic reticulum stress‐induced neuronal cell death triggered by expanded polyglutamine repeats, Genes Dev. 16, 1345‐1355. Novoa, I., Zhang, Y., Zeng, H., Jungreis, R., Harding, H.P. and Ron, D., 2003. Stress‐induced gene expression requires programmed recovery from translational repression, Embo j. 22, 1180‐ 1187. Ogg, S., Paradis, S., Gottlieb, S., Patterson, G.I., Lee, L., Tissenbaum, H.A. and Ruvkun, G., 1997. The Fork head transcription factor DAF‐16 transduces insulin‐like metabolic and longevity signals in C. elegans, Nature. 389, 994‐999. Oliver, J.D., Roderick, H.L., Llewellyn, D.H. and High, S., 1999. ERp57 functions as a subunit of specific complexes formed with the ER lectins calreticulin and calnexin, Mol Biol Cell. 10, 2573‐2582. Olmos, Y., Brosens, J.J. and Lam, E.W., 2011. Interplay between SIRT proteins and tumour suppressor transcription factors in chemotherapeutic resistance of cancer, Drug Resist Updat. 14, 35‐44. Ozcan, U., Ozcan, L., Yilmaz, E., Duvel, K., Sahin, M., Manning, B.D. and Hotamisligil, G.S., 2008. Loss of the tuberous sclerosis complex tumor suppressors triggers the unfolded protein response to regulate insulin signaling and apoptosis, Mol Cell. 29, 541‐551. Paik, J.H., Kollipara, R., Chu, G., Ji, H., Xiao, Y., Ding, Z., Miao, L., Tothova, Z., Horner, J.W., Carrasco, D.R., Jiang, S., Gilliland, D.G., Chin, L., Wong, W.H., Castrillon, D.H. and DePinho, R.A., 2007. FoxOs are lineage‐restricted redundant tumor suppressors and regulate endothelial cell homeostasis, Cell. 128, 309‐323. Papandreou, I., Denko, N.C., Olson, M., Van Melckebeke, H., Lust, S., Tam, A., Solow‐Cordero, D.E., Bouley, D.M., Offner, F., Niwa, M. and Koong, A.C., 2010. Identification of an Ire1alpha endonuclease specific inhibitor with cytotoxic activity against human multiple myeloma, Blood. 117, 1311‐1314. Paraiso, K.H., Haarberg, H.E., Wood, E., Rebecca, V.W., Chen, Y.A., Xiang, Y., Ribas, A., Lo, R.S., Weber, J.S., Sondak, V.K., John, J.K., Sarnaik, A.A., Koomen, J.M. and Smalley, K.S., 2012. The HSP90 inhibitor XL888 overcomes BRAF inhibitor resistance mediated through diverse mechanisms, Clin Cancer Res. 18, 2502‐2514. Peck, B., Chen, C.‐Y., Ho, K.‐K., Di Fruscia, P., Myatt, S.S., Coombes, R.C., Fuchter, M.J., Hsiao, C.‐D. and Lam, E.W.‐F., 2010. SIRT Inhibitors Induce Cell Death and p53 Acetylation through Targeting Both SIRT1 and SIRT2, Molecular Cancer Therapeutics. 9, 844‐855. Plemper, R.K., Bohmler, S., Bordallo, J., Sommer, T. and Wolf, D.H., 1997. Mutant analysis links the translocon and BiP to retrograde protein transport for ER degradation, Nature. 388, 891‐895. Pytel, D., Gao, Y., Mackiewicz, K., Katlinskaya, Y.V., Staschke, K.A., Paredes, M.C., Yoshida, A., Qie, S., Zhang, G., Chajewski, O.S., Wu, L., Majsterek, I., Herlyn, M., Fuchs, S.Y. and Diehl, J.A., 2016. PERK Is a Haploinsufficient Tumor Suppressor: Gene Dose Determines Tumor‐Suppressive Versus Tumor Promoting Properties of PERK in Melanoma, PLoS Genet. 12, e1006518. Pytel, D., Majsterek, I. and Diehl, J.A., 2016. Tumor progression and the different faces of the PERK kinase, Oncogene. 35, 1207‐1215. Qin, L., Wang, Z., Tao, L. and Wang, Y., 2010. ER stress negatively regulates AKT/TSC/mTOR pathway to enhance autophagy, Autophagy. 6, 239‐247. Rashid, H.‐O., Yadav, R.K., Kim, H.‐R. and Chae, H.‐J., 2015. ER stress: Autophagy induction, inhibition and selection, Autophagy. 11, 1956‐1977.

AC C

1058 1059 1060 1061 1062 1063 1064 1065 1066 1067 1068 1069 1070 1071 1072 1073 1074 1075 1076 1077 1078 1079 1080 1081 1082 1083 1084 1085 1086 1087 1088 1089 1090 1091 1092 1093 1094 1095 1096 1097 1098 1099 1100 1101 1102 1103 1104 1105 1106 1107 1108

30

ACCEPTED MANUSCRIPT

EP

TE D

M AN U

SC

RI PT

Rashid, H.O., Yadav, R.K., Kim, H.R. and Chae, H.J., 2015. ER stress: Autophagy induction, inhibition and selection, Autophagy. 11, 1956‐1977. Ri, M., Tashiro, E., Oikawa, D., Shinjo, S., Tokuda, M., Yokouchi, Y., Narita, T., Masaki, A., Ito, A., Ding, J., Kusumoto, S., Ishida, T., Komatsu, H., Shiotsu, Y., Ueda, R., Iwawaki, T., Imoto, M. and Iida, S., 2012. Identification of Toyocamycin, an agent cytotoxic for multiple myeloma cells, as a potent inhibitor of ER stress‐induced XBP1 mRNA splicing, Blood Cancer J. 2, e79. Rodriguez, D.A., Zamorano, S., Lisbona, F., Rojas‐Rivera, D., Urra, H., Cubillos‐Ruiz, J.R., Armisen, R., Henriquez, D.R., H Cheng, E., Letek, M., Vaisar, T., Irrazabal, T., Gonzalez‐Billault, C., Letai, A., Pimentel‐Muiños, F.X., Kroemer, G. and Hetz, C., 2012. BH3‐only proteins are part of a regulatory network that control the sustained signalling of the unfolded protein response sensor IRE1α, The EMBO Journal. 31, 2322‐2335. Romero‐Ramirez, L., Cao, H., Nelson, D., Hammond, E., Lee, A.H., Yoshida, H., Mori, K., Glimcher, L.H., Denko, N.C., Giaccia, A.J., Le, Q.T. and Koong, A.C., 2004. XBP1 Is Essential for Survival under Hypoxic Conditions and Is Required for Tumor Growth, Cancer Research. 64, 5943‐ 5947. Ron, D. and Walter, P., 2007. Signal integration in the endoplasmic reticulum unfolded protein response, Nat Rev Mol Cell Biol. 8, 519‐529. Rouschop, K.M.A., van den Beucken, T., Dubois, L., Niessen, H., Bussink, J., Savelkouls, K., Keulers, T., Mujcic, H., Landuyt, W., Voncken, J.W., Lambin, P., van der Kogel, A.J., Koritzinsky, M. and Wouters, B.G., 2010. The unfolded protein response protects human tumor cells during hypoxia through regulation of the autophagy genes MAP1LC3B and ATG5, Journal of Clinical Investigation. 120, 127‐141. Safra, M., Ben‐Hamo, S., Kenyon, C. and Henis‐Korenblit, S., 2013. The ire‐1 ER stress‐response pathway is required for normal secretory‐protein metabolism in C. elegans, J Cell Sci. 126, 4136‐4146. Safra, M., Fickentscher, R., Levi‐Ferber, M., Danino, Y.M., Haviv‐Chesner, A., Hansen, M., Juven‐ Gershon, T., Weiss, M. and Henis‐Korenblit, S., 2014. The FOXO transcription factor DAF‐16 bypasses ire‐1 requirement to promote endoplasmic reticulum homeostasis, Cell Metab. 20, 870‐881. Sasagawa, Y., Yamanaka, K. and Ogura, T., 2007. ER E3 ubiquitin ligase HRD‐1 and its specific partner chaperone BiP play important roles in ERAD and developmental growth in Caenorhabditis elegans, Genes Cells. 12, 1063‐1073. Schaheen, B., Dang, H. and Fares, H., 2009. Derlin‐dependent accumulation of integral membrane proteins at cell surfaces, J Cell Sci. 122, 2228‐2239. Sevier, C.S. and Kaiser, C.A., 2008. Ero1 and redox homeostasis in the endoplasmic reticulum, Biochim Biophys Acta. 1783, 549‐556. Shen, J., Chen, X., Hendershot, L. and Prywes, R., 2002. ER stress regulation of ATF6 localization by dissociation of BiP/GRP78 binding and unmasking of Golgi localization signals, Dev Cell. 3, 99‐111. Shen, X., Ellis, R.E., Lee, K., Liu, C.Y., Yang, K., Solomon, A., Yoshida, H., Morimoto, R., Kurnit, D.M., Mori, K. and Kaufman, R.J., 2001. Complementary signaling pathways regulate the unfolded protein response and are required for C. elegans development, Cell. 107, 893‐903. Shen, X., Ellis, R.E., Sakaki, K. and Kaufman, R.J., 2005. Genetic interactions due to constitutive and inducible gene regulation mediated by the unfolded protein response in C. elegans, PLoS Genet. 1, e37. Shuda, M., Kondoh, N., Imazeki, N., Tanaka, K., Okada, T., Mori, K., Hada, A., Arai, M., Wakatsuki, T., Matsubara, O., Yamamoto, N. and Yamamoto, M., 2003. Activation of the ATF6, XBP1 and grp78 genes in human hepatocellular carcinoma: a possible involvement of the ER stress pathway in hepatocarcinogenesis, Journal of Hepatology. 38, 605‐614. Sonneville, R. and Gonczy, P., 2004. Zyg‐11 and cul‐2 regulate progression through meiosis II and polarity establishment in C. elegans, Development. 131, 3527‐3543.

AC C

1109 1110 1111 1112 1113 1114 1115 1116 1117 1118 1119 1120 1121 1122 1123 1124 1125 1126 1127 1128 1129 1130 1131 1132 1133 1134 1135 1136 1137 1138 1139 1140 1141 1142 1143 1144 1145 1146 1147 1148 1149 1150 1151 1152 1153 1154 1155 1156 1157 1158 1159

31

ACCEPTED MANUSCRIPT

EP

TE D

M AN U

SC

RI PT

Spiotto, M.T., Banh, A., Papandreou, I., Cao, H., Galvez, M.G., Gurtner, G.C., Denko, N.C., Le, Q.T. and Koong, A.C., 2009. Imaging the Unfolded Protein Response in Primary Tumors Reveals Microenvironments with Metabolic Variations that Predict Tumor Growth, Cancer Research. 70, 78‐88. Strappazzon, F., Vietri‐Rudan, M., Campello, S., Nazio, F., Florenzano, F., Fimia, G.M., Piacentini, M., Levine, B. and Cecconi, F., 2011. Mitochondrial BCL‐2 inhibits AMBRA1‐induced autophagy, The EMBO Journal. 30, 1195‐1208. Sunters, A., Fernandez de Mattos, S., Stahl, M., Brosens, J.J., Zoumpoulidou, G., Saunders, C.A., Coffer, P.J., Medema, R.H., Coombes, R.C. and Lam, E.W., 2003. FoxO3a transcriptional regulation of Bim controls apoptosis in paclitaxel‐treated breast cancer cell lines, J Biol Chem. 278, 49795‐49805. Sunters, A., Madureira, P.A., Pomeranz, K.M., Aubert, M., Brosens, J.J., Cook, S.J., Burgering, B.M., Coombes, R.C. and Lam, E.W., 2006. Paclitaxel‐induced nuclear translocation of FOXO3a in breast cancer cells is mediated by c‐Jun NH2‐terminal kinase and Akt, Cancer Res. 66, 212‐ 220. Tatham, M.H., Geoffroy, M.C., Shen, L., Plechanovova, A., Hattersley, N., Jaffray, E.G., Palvimo, J.J. and Hay, R.T., 2008. RNF4 is a poly‐SUMO‐specific E3 ubiquitin ligase required for arsenic‐ induced PML degradation, Nat Cell Biol. 10, 538‐546. Teske, B.F., Wek, S.A., Bunpo, P., Cundiff, J.K., McClintick, J.N., Anthony, T.G. and Wek, R.C., 2011. The eIF2 kinase PERK and the integrated stress response facilitate activation of ATF6 during endoplasmic reticulum stress, Mol Biol Cell. 22, 4390‐4405. Thorpe, J.A. and Schwarze, S.R., 2009. IRE1α controls cyclin A1 expression and promotes cell proliferation through XBP‐1, Cell Stress and Chaperones. 15, 497‐508. Tissenbaum, H.A. and Guarente, L., 2001. Increased dosage of a sir‐2 gene extends lifespan in Caenorhabditis elegans, Nature. 410, 227‐230. Totani, K., Ihara, Y., Tsujimoto, T., Matsuo, I. and Ito, Y., 2009. The recognition motif of the glycoprotein‐folding sensor enzyme UDP‐Glc:glycoprotein glucosyltransferase, Biochemistry. 48, 2933‐2940. Tothova, Z., Kollipara, R., Huntly, B.J., Lee, B.H., Castrillon, D.H., Cullen, D.E., McDowell, E.P., Lazo‐ Kallanian, S., Williams, I.R., Sears, C., Armstrong, S.A., Passegue, E., DePinho, R.A. and Gilliland, D.G., 2007. FoxOs are critical mediators of hematopoietic stem cell resistance to physiologic oxidative stress, Cell. 128, 325‐339. Travers, K.J., Patil, C.K., Wodicka, L., Lockhart, D.J., Weissman, J.S. and Walter, P., 2000. Functional and genomic analyses reveal an essential coordination between the unfolded protein response and ER‐associated degradation, Cell. 101, 249‐258. Tullet, J.M., Hertweck, M., An, J.H., Baker, J., Hwang, J.Y., Liu, S., Oliveira, R.P., Baumeister, R. and Blackwell, T.K., 2008. Direct inhibition of the longevity‐promoting factor SKN‐1 by insulin‐like signaling in C. elegans, Cell. 132, 1025‐1038. Urano, F., Calfon, M., Yoneda, T., Yun, C., Kiraly, M., Clark, S.G. and Ron, D., 2002. A survival pathway for Caenorhabditis elegans with a blocked unfolded protein response, J Cell Biol. 158, 639‐ 646. Urano, F., Wang, X., Bertolotti, A., Zhang, Y., Chung, P., Harding, H.P. and Ron, D., 2000. Coupling of stress in the ER to activation of JNK protein kinases by transmembrane protein kinase IRE1, Science. 287, 664‐666. van der Horst, A., de Vries‐Smits, A.M., Brenkman, A.B., van Triest, M.H., van den Broek, N., Colland, F., Maurice, M.M. and Burgering, B.M., 2006. FOXO4 transcriptional activity is regulated by monoubiquitination and USP7/HAUSP, Nat Cell Biol. 8, 1064‐1073. Vandewynckel, Y.P., Laukens, D., Geerts, A., Bogaerts, E., Paridaens, A., Verhelst, X., Janssens, S., Heindryckx, F. and Van Vlierberghe, H., 2013. The paradox of the unfolded protein response in cancer, Anticancer Res. 33, 4683‐4694.

AC C

1160 1161 1162 1163 1164 1165 1166 1167 1168 1169 1170 1171 1172 1173 1174 1175 1176 1177 1178 1179 1180 1181 1182 1183 1184 1185 1186 1187 1188 1189 1190 1191 1192 1193 1194 1195 1196 1197 1198 1199 1200 1201 1202 1203 1204 1205 1206 1207 1208 1209

32

ACCEPTED MANUSCRIPT

EP

TE D

M AN U

SC

RI PT

Vattem, K.M. and Wek, R.C., 2004. Reinitiation involving upstream ORFs regulates ATF4 mRNA translation in mammalian cells, Proceedings of the National Academy of Sciences. 101, 11269‐11274. Verfaillie, T., Salazar, M., Velasco, G. and Agostinis, P., 2010. Linking ER Stress to Autophagy: Potential Implications for Cancer Therapy, International Journal of Cell Biology. 2010, 1‐19. Volkmann, K., Lucas, J.L., Vuga, D., Wang, X., Brumm, D., Stiles, C., Kriebel, D., Der‐Sarkissian, A., Krishnan, K., Schweitzer, C., Liu, Z., Malyankar, U.M., Chiovitti, D., Canny, M., Durocher, D., Sicheri, F. and Patterson, J.B., 2011. Potent and Selective Inhibitors of the Inositol‐requiring Enzyme 1 Endoribonuclease, Journal of Biological Chemistry. 286, 12743‐12755. Wada, I., Imai, S., Kai, M., Sakane, F. and Kanoh, H., 1995. Chaperone function of calreticulin when expressed in the endoplasmic reticulum as the membrane‐anchored and soluble forms, J Biol Chem. 270, 20298‐20304. Wang, F., Chan, C.H., Chen, K., Guan, X., Lin, H.K. and Tong, Q., 2011. Deacetylation of FOXO3 by SIRT1 or SIRT2 leads to Skp2‐mediated FOXO3 ubiquitination and degradation, Oncogene. In Press, doi: 10.1038/onc.2011.1347. Wang, M. and Kaufman, R.J., 2014. The impact of the endoplasmic reticulum protein‐folding environment on cancer development, Nat Rev Cancer. 14, 581‐597. Wang, M. and Kaufman, R.J., 2016. Protein misfolding in the endoplasmic reticulum as a conduit to human disease, Nature. 529, 326‐335. Wolff, S. and Dillin, A., 2006. The trifecta of aging in Caenorhabditis elegans, Exp Gerontol. 41, 894‐ 903. Yamaguchi, H. and Wang, H.G., 2004. CHOP is involved in endoplasmic reticulum stress‐induced apoptosis by enhancing DR5 expression in human carcinoma cells, J Biol Chem. 279, 45495‐ 45502. Yamamoto, S., Tomita, Y., Nakamori, S., Hoshida, Y., Nagano, H., Dono, K., Umeshita, K., Sakon, M., Monden, M. and Aozasa, K., 2003. Elevated expression of valosin‐containing protein (p97) in hepatocellular carcinoma is correlated with increased incidence of tumor recurrence, J Clin Oncol. 21, 447‐452. Yamanaka, A., Yada, M., Imaki, H., Koga, M., Ohshima, Y. and Nakayama, K., 2002. Multiple Skp1‐ related proteins in Caenorhabditis elegans: diverse patterns of interaction with Cullins and F‐ box proteins, Curr Biol. 12, 267‐275. Yan, W., Frank, C.L., Korth, M.J., Sopher, B.L., Novoa, I., Ron, D. and Katze, M.G., 2002. Control of PERK eIF2alpha kinase activity by the endoplasmic reticulum stress‐induced molecular chaperone P58IPK, Proc Natl Acad Sci U S A. 99, 15920‐15925. Yang, Z., Xu, Y., Xu, L., Maccauro, G., Rossi, B., Chen, Y., Li, H., Zhang, J., Sun, H., Yang, Y., Xu, D. and Liu, X., 2013. Regulation of Autophagy Via PERK‐eIF2α Effectively Relieve the Radiation Myelitis Induced by Iodine‐125, PLoS ONE. 8, e76819. Yoneda, T., Imaizumi, K., Oono, K., Yui, D., Gomi, F., Katayama, T. and Tohyama, M., 2001. Activation of caspase‐12, an endoplastic reticulum (ER) resident caspase, through tumor necrosis factor receptor‐associated factor 2‐dependent mechanism in response to the ER stress, J Biol Chem. 276, 13935‐13940. Yoshida, H., Matsui, T., Yamamoto, A., Okada, T. and Mori, K., 2001. XBP1 mRNA Is Induced by ATF6 and Spliced by IRE1 in Response to ER Stress to Produce a Highly Active Transcription Factor, Cell. 107, 881‐891. Yoshida, H., Okada, T., Haze, K., Yanagi, H., Yura, T., Negishi, M. and Mori, K., 2000. ATF6 activated by proteolysis binds in the presence of NF‐Y (CBF) directly to the cis‐acting element responsible for the mammalian unfolded protein response, Mol Cell Biol. 20, 6755‐6767. Yung, H.W., Charnock‐Jones, D.S. and Burton, G.J., 2011. Regulation of AKT Phosphorylation at Ser473 and Thr308 by Endoplasmic Reticulum Stress Modulates Substrate Specificity in a Severity Dependent Manner, PLoS ONE. 6, e17894.

AC C

1210 1211 1212 1213 1214 1215 1216 1217 1218 1219 1220 1221 1222 1223 1224 1225 1226 1227 1228 1229 1230 1231 1232 1233 1234 1235 1236 1237 1238 1239 1240 1241 1242 1243 1244 1245 1246 1247 1248 1249 1250 1251 1252 1253 1254 1255 1256 1257 1258 1259

33

ACCEPTED MANUSCRIPT

RI PT

Zeng, L., Lu, M., Mori, K., Luo, S., Lee, A.S., Zhu, Y. and Shyy, J.Y.J., 2004. ATF6 modulates SREBP2‐ mediated lipogenesis, EMBO J. 23, 950‐958. Zhang, W., Hietakangas, V., Wee, S., Lim, S.C., Gunaratne, J. and Cohen, S.M., 2013. ER stress potentiates insulin resistance through PERK‐mediated FOXO phosphorylation, Genes Dev. 27, 441‐449. Zhang, W., Neo, S.P., Gunaratne, J., Poulsen, A., Boping, L., Ong, E.H., Sangthongpitag, K., Pendharkar, V., Hill, J. and Cohen, S.M., 2015. Feedback regulation on PTEN/AKT pathway by the ER stress kinase PERK mediated by interaction with the Vault complex, Cell Signal. 27, 436‐442. Zhao, Y., Li, X., Cai, M.Y., Ma, K., Yang, J., Zhou, J., Fu, W., Wei, F.Z., Wang, L., Xie, D. and Zhu, W.G., 2013. XBP‐1u suppresses autophagy by promoting the degradation of FoxO1 in cancer cells, Cell Res. 23, 491‐507. Zhou, Y., Lee, J., Reno, C.M., Sun, C., Park, S.W., Chung, J., Lee, J., Fisher, S.J., White, M.F., Biddinger, S.B. and Ozcan, U., 2011. Regulation of glucose homeostasis through a XBP‐1–FoxO1 interaction, Nature Medicine. 17, 356‐365. Zona, S., Bella, L., Burton, M.J., Nestal de Moraes, G. and Lam, E.W., 2014. FOXM1: an emerging master regulator of DNA damage response and genotoxic agent resistance, Biochim Biophys Acta. 1839, 1316‐1322.

SC

1260 1261 1262 1263 1264 1265 1266 1267 1268 1269 1270 1271 1272 1273 1274 1275 1276 1277

M AN U

1278

AC C

EP

TE D

1279

34

ACCEPTED MANUSCRIPT Figure Legends

1281

Figure.1 Accumulation of misfolded protein induces UPR activity and auto-

1282

phosphorylation of PERK. Activation of PERK attenuates the global translation by

1283

phosphorylating the α subunit of eIF2 to decrease the load of protein folding in ER. As a

1284

result, the specifically translated ATF4 will induce transcription of pro-survival genes for

1285

antioxidant response, amino acid synthesis, ER Chaperones, autophagy and combine with

1286

FOXO to regulate glucose level under acute stress. Upon chronic stress, ATF4 transcribes

1287

CHOP which in turn will induce expression of ROS synthesis and pro-apoptotic genes.

1288

Moreover, CHOP will promote transcription of GADD34 to dephosphorylate eIF2α and

1289

restore the global mRNA translation. (ATF4 = Activating transcription factor 4, CHOP =

1290

CCAAT/enhancer-binding (C/EBP) homologous protein, eIF2 = Eukaryotic translation

1291

initiation factor-2, ER = Endoplasmic reticulum, GADD34 = Growth arrest and DNA-damage-

1292

inducible protein 34, PERK = PKR-like endoplasmic reticulum kinase, ROS = Reactive

1293

oxygen species, UPR = Unfolded protein response).

M AN U

SC

RI PT

1280

1294

Figure. 2 ER stress signalling through IRE1α and ATF6. Upon accumulation of misfolded

1296

proteins, BiP disassociates from IRE1α and ATF6. IRE1α dimerizes and induces its

1297

endoribonuclease activity. Under survival condition, introns on XBP1 mRNA will be spliced

1298

to form XBP1s that works as transcription factors to induce the expression of genes for ER

1299

chaperones, lipid synthesis and ERAD process. ATF6 will be cleaved at Golgi by SP1 and

1300

SP2 proteases to form transcription factor ATF6α which increases the transcription of genes

1301

for XBP1, ER chaperones and ERAD process. However, IRE1α activation causes

1302

degradation of mRNA by the RIDD process to decrease protein folding and apoptosis.

1303

Moreover, IRE1α mediates phosphorylation of JNK which in turn induces FOXO3 to send

1304

cells for either autophagy or apoptosis. (ATF6 = Activation factor 6, BiP = Immunoglobulin

1305

heavy-chain binding protein, ER = Endoplasmic reticulum, ERAD = ER-associated

1306

degradation, GRP94 = 94 kDa glucose-regulated protein, FOXO3 = Forkhead box O3,

1307

IRE1α = Inositol requiring enzyme 1α, JNK = c-JUN N-terminal kinase, RIDD = IRE1α-

1308

dependent decay, SP1 = Site-1 proteases, SP2 = Site-2 proteases, XBP1 = X-box binding

1309

protein 1).

EP

AC C

1310

TE D

1295

1311

Figure.3 The role of PERK in FOXO3 regulation and ER stress signalling.

1312

regulates FOXO3 directly by phosphorylating five sites, including S261, S298, S301, S303

1313

and S311, which induces its activity and translocation to nucleus. However, PERK inhibits

1314

FOXO3 activity to induce insulin resistance, obesity, adipocytic differentiation and cancer

1315

progression through mTOR/AKT. PERK induces mTOR to phosphorylate AKT on S473

1316

which causes relocation of FOXO3 to cytoplasm and degradation. Moreover, CHOP,

35

PERK

ACCEPTED MANUSCRIPT downstream target of PERK, cooperates with FOXO3 to induce pre-apoptotic genes

1318

expression. Furthermore, CHOP mediates ROS activity by induce ERO1α to activate JNK

1319

which in turn phosphorylates FOXO3 and induces apoptosis. (ATF4 = Activating

1320

Transcription Factor 4, AKT = Protein Kinase B, CHOP = CCAAT/enhancer-binding (C/EBP)

1321

homologous protein, eIF2 = eukaryotic translation initiation factor-2, ER = endoplasmic

1322

reticulum, ERO1α = ER oxidoreductin 1α, FOXO3 = Forkhead box O3, JNK = c-Jun N-

1323

terminal kinases, mTOR = mechanistic/ mammalian target of rapamycin, PERK = PKR-like

1324

endoplasmic reticulum kinase, ROS = Reactive oxygen species).

1325

RI PT

1317

Table 1 Functional human orthologues of C. elegans ER stress-related genes and

1327

their functions and relationship with Daf-16/FOXO. A role in the regulations of ER stress

1328

response to promote ER stress resistance and longevity. In C. elegans, ER stress

1329

responses, particularly the unfolded protein response (UPR), are mainly dependent on the

1330

Ire-1/Xbp-1 pathway. In fact, the downstream gene targets of the ire-1/xbp-1 pathway

1331

accounts for the majority of UPR genes which are implicated in protein folding and

1332

endoplasmic reticulum-associated protein degradation (ERAD), such as cul-1, cul-2, skr,

1333

and cup-2. Interestingly, xbp-1 was found to collaborate with daf-16 to activate transcription

1334

of these downstream targets. Studies of C. elegans also showed that pek-1 acts

1335

synergistically with atf-6 to maintain ER homeostasis as an alternative mechanism to the ire-

1336

1/xbp-1 pathway. Furthermore, pek-1 and atf-6 are both found to interact with daf-16 during

1337

ER stress response, suggesting a role of daf-16 in maintain ER homeostasis that is

1338

independent of the ire-1/xbp-1 signaling pathway

1341

M AN U

TE D

EP

1340

AC C

1339

SC

1326

36

ACCEPTED MANUSCRIPT Highlights

RI PT

SC M AN U TE D



EP

• •

A comprehensive analysis of the main ER stress players and signalling pathways. New insights into the ER stress activation and its role in cancer initiation and progression. FOXO3 as a new key molecule within the ER stress network. Regulation of FOXO3 by PERK and CHOP and its implications in cancer progression and resistance. Significance of FOXO3 in the UPR signalling and new strategies to overcome drug resistance.

AC C

• •