Author Manuscript Published OnlineFirst on June 12, 2015; DOI: 10.1158/1078-0432.CCR-14-3163 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

1

Autocrine activation of CHRM3 promotes prostate cancer growth and

2

castration resistance via CaM/CaMKK-mediated phosphorylation of Akt

3

Naitao Wang, Ming Yao, Jin Xu, Yizhou Quan, Kaiqing Zhang, Ru Yang* and

4

Wei-Qiang Gao*

5 6

Authors’ Affiliations:

7

State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem

8

Cell Research Center, Ren Ji Hospital, School of Biomedical Engineering, Shanghai

9

Jiao Tong University,Shanghai, China.

10 11

Running Title: Autocrine cholinergic signaling in prostate cancer

12 13

Financial support: The study is supported by funds to Wei-Qiang Gao from the

14

Chinese Ministry of Science and Technology (2012CB966800, 2013CB945600 and

15

2012CB967903), the National Natural Science Foundation of China (81130038 and

16

81372189), the Science and Technology Commission of Shanghai Municipality

17

(Pujiang program), the Shanghai Education Committee Key Discipline and Specialty

18

Foundation (J50208), the Shanghai Health Bureau Key Discipline and Specialty

19

Foundation and the KC Wong foundation.

20

1 Downloaded from clincancerres.aacrjournals.org on July 23, 2015. © 2015 American Association for Cancer Research.

Author Manuscript Published OnlineFirst on June 12, 2015; DOI: 10.1158/1078-0432.CCR-14-3163 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

21

*Corresponding Authors: Wei-Qiang Gao or Ru Yang, Ren Ji Hospital, School

22

of Medicine, Shanghai Jiao Tong University, Shanghai, China.

23

E-mail: [email protected] or [email protected]

24 25

The authors declare no potential conflicts of interest

26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41

2 Downloaded from clincancerres.aacrjournals.org on July 23, 2015. © 2015 American Association for Cancer Research.

Author Manuscript Published OnlineFirst on June 12, 2015; DOI: 10.1158/1078-0432.CCR-14-3163 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

42

Statement of translational relevance

43

This study aimed to elucidate the comprehensive roles of autocrine cholinergic

44

signaling in prostate cancer growth and castration resistance. Although androgen

45

deprivation is initially effective for the treatment of prostate cancer, patients

46

inevitably develop resistance to androgen deprivation therapy, called castration

47

resistance, which is the major cause of morbidity and mortality. To date, there is

48

no effective therapy available for the treatment of castration resistant prostate

49

cancer (CRPC). The present work demonstrates an autocrine activation of the

50

cholinergic system in prostate cancer. Importantly, a selective cholinergic

51

muscarinic receptor 3 (CHRM3) antagonist, darifenacin, effectively inhibits

52

prostate cancer growth and castration resistance both in vitro and in vivo,

53

suggesting a potential therapeutic application of selective CHRM3 antagonists in

54

the treatment of prostate cancer, including CRPC.

55 56 57 58 59 60 61 62 63 64 65 66

3 Downloaded from clincancerres.aacrjournals.org on July 23, 2015. © 2015 American Association for Cancer Research.

Author Manuscript Published OnlineFirst on June 12, 2015; DOI: 10.1158/1078-0432.CCR-14-3163 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

67

Abstract

68

Purpose: Although a previous study reported nerve ending-derived acetylcholine

69

promoted

70

microenvironment of cancer cells, the present study aims to determine whether

71

there is autocrine cholinergic signaling in prostate epithelial cells that promotes

72

prostate cancer growth and castration resistance.

73

Experimental design: In this study, immunohistochemistry (IHC) was performed

74

to detect protein expression in mouse prostate tissue sections and human

75

prostate cancer tissue sections. Subcutaneously and orthotopically xenografted

76

tumor models were established to evaluate the functions of autocrine cholinergic

77

signaling in regulating prostate cancer growth and castration resistance. Western

78

blotting analysis was performed to assess the autocrine cholinergic signaling-

79

induced signaling pathway.

80

Results: We found the expression of choline acetyltransferase (ChAT), the

81

secretion of acetylcholine and the expression of CHRM3 in prostate epithelial

82

cells, supporting the presence of autocrine cholinergic signaling in the prostate

83

epithelium. In addition, we found that CHRM3 was up-regulated in clinical

84

prostate cancer tissues compared to adjacent non-cancer tissues. Over-

85

expression of CHRM3 or activation of CHRM3 by carbachol promoted cell

86

proliferation, migration and castration resistance. On the contrary, blockading

87

CHRM3 by shRNA or treatment with darifenacin inhibited prostate cancer growth

88

and castration resistance both in vitro and in vivo. Furthermore, we found that

89

autocrine cholinergic signaling caused calmodulin/calmodulin-dependent protein

90

kinase kinase (CaM/CaMKK)-mediated phosphorylation of Akt.

91

Conclusions: These findings suggest that blockade of CHRM3 may represent a

92

novel adjuvant therapy for CRPC.

prostate

cancer

invasion

and

metastasis

by

regulating

the

93 94

4 Downloaded from clincancerres.aacrjournals.org on July 23, 2015. © 2015 American Association for Cancer Research.

Author Manuscript Published OnlineFirst on June 12, 2015; DOI: 10.1158/1078-0432.CCR-14-3163 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

95

Introduction

96

Prostate cancer is the most common malignant disease in men in developed

97

countries (1). In 2014, 233,000 new cases of prostate cancer are expected to

98

occur in the United States, which accounts for 27% of incident cases in men (1).

99

Although androgen deprivation is initially effective, the patients inevitably

100

encounter the problem of resistance to androgen deprivation therapy, called

101

castration resistance (2). Discovery of novel strategies to suppress castration

102

resistance is especially helpful for the management of advanced prostate cancer.

103 104

Acetylcholine,

normally

released

from

nerve

endings,

is

a

classical

105

neurotransmitter in the central and peripheral nervous system. A recent study

106

reported the involvement of nerve ending-derived acetylcholine activated

107

cholinergic muscarinic receptor 1 (CHRM1) in mesenchymal cells in the tumor

108

microenvironment to promote prostate cancer invasion and metastasis (3).

109

However, besides nerve ending-derived acetylcholine, there is also wide-spread

110

synthesis of acetylcholine by a variety of non-neuronal cell types, including lung,

111

colon, airway and ovarian epithelial cells (4). Whether there is synthesis of

112

acetylcholine in prostate epithelial cells which might play an autocrine activation

113

role in promoting prostate cancer progression has not yet been studied.

114 115

Muscarinic receptors are G-protein coupled receptors consisting of five members,

116

CHRM1-CHRM5 (5). Activation of muscarinic receptors triggers Ca2+ influx,

117

which causes smooth muscle contraction and glandular secretion. In the 1990s,

118

muscarinic receptor subtypes were firstly defined as conditional oncogenes when

119

they were activated by acetylcholine in NIH-3T3 cells (6). Subsequently,

120

muscarinic receptors have been implicated to be involved in a few types of

121

epithelial cancers including colorectal cancer and small cell lung cancer (7,8). In

122

addition, activation of muscarinic receptors was reported to promote prostate

123

cancer cell proliferation in vitro (9). However, the mechanism and comprehensive

5 Downloaded from clincancerres.aacrjournals.org on July 23, 2015. © 2015 American Association for Cancer Research.

Author Manuscript Published OnlineFirst on June 12, 2015; DOI: 10.1158/1078-0432.CCR-14-3163 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

124

functions of muscarinic receptors in prostate cancer progression, particularly in

125

CRPC, has not been clearly elucidated.

126 127

In the present study, we found the presence of a functional cholinergic system in

128

prostate epithelial cells. An autocrine activation of CHRM3 that regulated prostate

129

cancer cell growth and castration resistance was observed. Blockade of CHRM3

130

via a specific antagonist or shRNAs could effectively inhibit prostate cancer

131

growth and castration resistance. The cholinergic signaling occurred via the

132

CaM/CaMKK-mediated phosphorylation of Akt. These findings suggest a

133

potential application of muscarinic receptor antagonists in the treatment of

134

prostate cancer.

135 136

Materials and methods

137

Cell culture

138

Human prostate cell lines used in this study: LNCaP, PNT1B (10), BPH1 (a kind

139

gift from Dr. Simon W. Hayward) (11), C4-2B, PC3, PC3-AR+ and PC3-luc (kind

140

gifts from Dr. Jianhua Wang at Shanghai Jiao Tong University School of Medicine)

141

(12,13). Cells were cultured in DMEM or RPMI-1640 supplemented with 10%

142

FBS and P/S (penicillin and streptomycin) at 37°C, 5% CO2. In the cell

143

proliferation assay, cell numbers were determined by hemocytometer.

144 145

Cell transfection

146

Two shRNA sequences for CHRM3 (5’-3’, AGCAGAGACAGTCGGTCATTT and

147

TCGGCAACATCCTGGTAATTG) and scrambled shRNA were cloned into the

148

lentiviral vector pUCTP. The cDNA of CHRM3 was subcloned into the lentiviral

149

expression vector EGH. Transfection efficiencies were determined at both the

150

protein and mRNA levels.

6 Downloaded from clincancerres.aacrjournals.org on July 23, 2015. © 2015 American Association for Cancer Research.

Author Manuscript Published OnlineFirst on June 12, 2015; DOI: 10.1158/1078-0432.CCR-14-3163 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

151 152

Cell migration assays

153

In the scratch tests, confluent monolayer cells were scraped with a pipette tip.

154

Cells were cultured in standard cell culture medium containing 10% FBS.

155

Photographs were taken immediately and at 24 hours after wounding. To rule out

156

the potential effects of cell proliferation on cell migration, we normalized the cell

157

migration distance with the total number of cells at 24 hours.

158

In the transwell assays, cells were first starved in serum-free medium for 24

159

hours. Ten thousand-50,000 cells in 100 μL serum free medium were seeded on

160

the top of a transwell chamber (8 μm pore size, 3422, Corning), 500 μL medium

161

containing 10% FBS was added in the bottom of 24-well plates to induce cell

162

migration. Twelve hours later, the culture inserts were fixed with 4%

163

paraformaldehyde and stained in 0.1% crystal violet for 10 minutes. Cells that

164

stayed on the top of the membrane were gently scraped by a cotton swab. To

165

eliminate the effects of cell proliferation on cell migration, we starved PNT1B cells

166

(Lenti-vector and Lenti-CHRM3 transfected PNT1B cells) and PC3 cells

167

(scramble, shRNA1 and shRNA2 transfected PC3 cells) in serum free medium

168

for 24 hours and replaced the medium with standard cell culture medium for 12

169

hours. We measured the cell numbers before and at 12 hours after the

170

replacement of the standard cell culture medium and calculated the cell number

171

changes in both PNT1B cells and PC3 cells (data not shown). Then, we

172

normalized the results in transwell assays with the total number of cells at 12

173

hours.

174 175

In vivo experiments

176

For the subcutaneously xenografted tumor models, 1,000,000 cells (50 μL cell

177

suspension + 50 μL matrigel) were injected subcutaneously into the flank regions

178

of 6-week-old male BALB/c nude mice. Tumor volume was measured once a

7 Downloaded from clincancerres.aacrjournals.org on July 23, 2015. © 2015 American Association for Cancer Research.

Author Manuscript Published OnlineFirst on June 12, 2015; DOI: 10.1158/1078-0432.CCR-14-3163 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

179

week (V=0.5ab2; a, the longest side; b, the shortest side). For orthotopical

180

implantation assays, 8-week-old male BALB/c nude mice were first castrated.

181

Two weeks later, 1,000,000 cells (25 μL cell suspension + 25 μL matrigel) were

182

injected into the anterior lobes of the prostate with a 31G insulin syringe.

183

Bioluminescent signal was induced by intraperitoneal injection of D-luciferin

184

(GoldBio, 1.5 mg D-luciferin per 10 g body weight in saline) and analyzed 10

185

minutes later using a Berthold imaging system. In some assays, darifenacin was

186

injected intraperitoneally at doses of 1 mg/kg/day and 5 mg/kg/day. Vehicle (PBS

187

mixed with DMSO at 1:1) was injected in the control group. Mice were fed in the

188

SPF grade animal facility of Ren Ji hospital with controlled temperature and

189

humidity. All animal studies were carried out following the guidelines of the Ren Ji

190

hospital institutional animal care and ethics committee.

191 192

Clinical prostate cancer samples

193

Paraffin embedded human prostate tissue array slides containing fifty-eight spots

194

(29 paired prostate cancer and adjacent non-cancerous tissues) (OD-CT-

195

UrPrt03-001) were purchased from Shanghai Outdo Biotech Ltd.. A primary

196

antibody against CHRM3 (Abbiotec) and HRP conjugated secondary antibodies

197

(Jackson ImmunoResearch) were applied. The immunostaining was visualized

198

with DAB (3, 3’-diaminobenzidine). Images were captured using a Leica DM2500

199

microscope under the same exposure conditions and analyzed with the Image-

200

pro Plus 6.0 software (Media Cybernetics).

201 202

Calcium influx detection

203

Cells were incubated with Fluo-4 AM (Invitrogen, 2 μM in Hank’s Balanced Salt

204

Solution, HBSS) for 60 minutes at 37°C and incubated for another 15 minutes in

205

fresh HBSS to allow de-esterification of intracellular AM esters. Acetylcholine and

206

carbachol were added to the culture medium to trigger a Ca2+ influx. For

8 Downloaded from clincancerres.aacrjournals.org on July 23, 2015. © 2015 American Association for Cancer Research.

Author Manuscript Published OnlineFirst on June 12, 2015; DOI: 10.1158/1078-0432.CCR-14-3163 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

207

antagonist studies, darifenacin was added 5 minutes before the stimulation of 10

208

μM carbachol. Images were taken with a Leica inverted microscope for 300

209

seconds at intervals of 6 seconds. Data were analyzed with the Image-pro Plus

210

6.0 software.

211 212

Western blot and immunoprecipitation

213

In western blotting assays, cells were lysed in a RIPA buffer (Thermo) with

214

proteinase inhibitors and phosphatase inhibitors (Roche). Total proteins were

215

measured by a BCA method (Thermo). Primary antibodies against GAPDH, E-

216

cadherin, N-cadherin, Vimentin (Epitomic), CaMKKα (Santa Cruz), CHRM3

217

(Abbiotec), Flag (Sigma), Slug, Akt, pAkt-Ser473, pAkt-Thr308 (CST) and HRP

218

conjugated secondary antibodies were applied. Immunoblots were visualized

219

with an ECL blotting detection kit (Thermo). In the immunoprecipitation assay,

220

cell lysates were incubated with the rabbit Akt antibody or control mouse IgG

221

antibody at 4°C overnight. Protein A-agarose beads (Roche) were added, and

222

lysates were further incubated for 1 h at room temperature. Beads were

223

precipitated by centrifugation at 5,000 rpm for 3 minutes and boiled in SDS-page

224

loading buffer for 5 minutes. The samples were then detected according to

225

standard western blotting procedures.

226 227

Immunofluorescent staining

228

Immunofluorescent staining was performed as previously described (14).

229

Antibodies used in this study included Tuj-1 (Sigma), ChAT (Millipore), CHRM3

230

(Abbiotec), Ki67, E-cadherin, N-cadherin, Vimentin (Epitomic) and Cleaved-

231

caspase3 (CST). The TUNEL apoptosis detection system (Promega) was used to

232

detect apoptotic cells in mouse xenografted tumor sections. Images were taken

233

using a Leica DM2500 microscope.

234

9 Downloaded from clincancerres.aacrjournals.org on July 23, 2015. © 2015 American Association for Cancer Research.

Author Manuscript Published OnlineFirst on June 12, 2015; DOI: 10.1158/1078-0432.CCR-14-3163 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

235

Real-time PCR

236

Total RNA was obtained and reverse-transcripted to cDNA by the RNeasy plus

237

Mini Kit and the QuantiTect RT Kit (Qiagen). SYBR (Roche) real-time PCR was

238

performed on the 7900HT machine (ABI). The sequences of the primers used in

239

this study are listed in Supplementary Table S1.

240 241

Statistical analysis

242

Data in this study are expressed as the means ± SEM. Immunostaining densities

243

of CHRM3 in matched human prostate cancerous and adjacent non-cancerous

244

tissues were compared by paired Student’s t-test. Cell proliferation, real-time

245

PCR and in vivo tumor xenograft growths were analyzed using a non-parametric

246

Student’s t-test. Ki67, TUNEL and cleaved-caspase3 positive cells were counted

247

in at least three randomly-selected visual fields and analyzed using a

248

nonparametric Student’s t-test. The Kaplan–Meier log-rank test was used for

249

analysis of mouse survival data. Data were analyzed with GraphPad Prism 5

250

software (GraphPad Software). Statistical significance was defined as * P < 0.05,

251

** P < 0.01, *** P < 0.001.

252 253 254 255 256 257 258 259

10 Downloaded from clincancerres.aacrjournals.org on July 23, 2015. © 2015 American Association for Cancer Research.

Author Manuscript Published OnlineFirst on June 12, 2015; DOI: 10.1158/1078-0432.CCR-14-3163 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

260

Results

261

Presence of functional autocrine cholinergic signaling in the prostate

262

epithelium

263

Autonomic nerves were reported to regulate prostate cancer metastasis by

264

regulating the microenvironment of epithelial cancer cells (3). Immunostaining of

265

mouse prostate sections confirmed dense innervation of Tuj-1 immunoreactive

266

nerve fibers; however, few, if any, fibers could be observed inside the epithelium

267

(Fig.

268

immunoreactivity of ChAT, which is the enzyme necessary for acetylcholine

269

synthesis (Fig. 1B). Importantly, ChAT immunoreactivity in human prostate

270

sections also showed the expression of ChAT in the epithelium (Fig. 1C). In

271

addition, we detected the secretion of acetylcholine from both cancerous and

272

non-cancerous human prostate epithelial cells, which are free of any neuronal

273

innervations (Fig. 1D). These data demonstrate clearly that there is endogenous

274

production and secretion of acetylcholine from prostate cancerous and non-

275

cancerous epithelial cells.

1A).

Instead,

mouse

prostate

epithelial

cells

showed

strong

276 277

A complete, functional cholinergic loop requires the presence of not only

278

acetylcholine but also muscarinic receptors. To determine the expression

279

patterns of muscarinic receptors in the prostate, we profiled gene expression in

280

the Oncomine database and GEO Profiles database. Of five muscarinic

281

cholinergic receptors, only CHRM3 was significantly elevated in prostate cancer

282

samples compared to non-cancer samples (Supplementary Fig. S1). To confirm

283

the Oncomine and GEO data analysis, we performed immunohistochemistry of

284

CHRM3 in the human prostate cancer tissue array and found that CHRM3 was

285

mainly expressed in the prostate epithelium rather than in the mesenchyme (Fig.

286

1E). Notably, CHRM3 was significantly up-regulated in cancer tissues compared

287

to their matched, adjacent non-cancer tissues (Fig. 1F).

288

11 Downloaded from clincancerres.aacrjournals.org on July 23, 2015. © 2015 American Association for Cancer Research.

Author Manuscript Published OnlineFirst on June 12, 2015; DOI: 10.1158/1078-0432.CCR-14-3163 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

289

To determine whether the autocrine acetylcholine could activate muscarinic

290

receptors in epithelial cells, we performed experiments to measure Ca2+ influx,

291

which is a direct indicator of muscarinic receptor activation. We measured Ca2+

292

influx with Fluo-4 AM, a commonly used fluorescent Ca2+ influx indicator, and

293

found that both acetylcholine and its stable analogue carbachol could induce

294

Ca2+ influx. CHRM3-specific antagonist darifenacin could largely reduce Ca2+

295

influx induced by carbachol (Fig. 1G). In addition, conditioned medium from PC3

296

cells also triggered Ca2+ influx, suggesting the occurrence of secretion of

297

endogenous non-nerve ending-derived acetylcholine (Fig. 1H). Importantly, the

298

Ca2+ influx induced by endogenous acetylcholine could be effectively blocked by

299

the CHRM3 specific inhibitor (Fig. 1H). Taken together, these findings show the

300

presence of functional autocrine cholinergic signaling in prostate epithelium.

301 302

Activation of CHRM3 by endogenous acetylcholine promotes prostate

303

cancer growth

304

To evaluate the role of autocrine cholinergic signaling in prostate cancer growth,

305

we over-expressed CHRM3 in non-tumorigenic PNT1B cells. Over-expression of

306

CHRM3 in PNT1B cells promoted cell growth as time proceeded (Fig. 2A),

307

implicating that over-expressed CHRM3 could be activated by endogenous

308

acetylcholine. In contrast, the knock-down of CHRM3 by shRNA in PC3 cells

309

inhibited cell growth (Fig. 2C). The lentiviral transfection efficiencies were

310

analyzed by western blot (Fig. 2B and 2D). In addition, we treated PC3 cells with

311

carbachol (a stable agonist of muscarinic receptors) and darifenacin (a selective

312

antagonist of CHRM3). While carbachol promoted the proliferation of PC3 and

313

22Rv1 cells, blockade of CHRM3 by darifenacin could effectively reduce cell

314

proliferation (Fig. 2E, F).

315 316

To extend our in vitro studies to an in vivo setting, we implanted CHRM3 knock-

317

down PC3 cells subcutaneously in BALB/c nude mice. We found that knock-

12 Downloaded from clincancerres.aacrjournals.org on July 23, 2015. © 2015 American Association for Cancer Research.

Author Manuscript Published OnlineFirst on June 12, 2015; DOI: 10.1158/1078-0432.CCR-14-3163 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

318

down of CHRM3 reduced tumor growth in vivo (Fig. 2G). Consistently, CHRM3-

319

specific antagonist darifenacin also inhibited the growth of xenografted PC3 and

320

22Rv1 cells (Fig. 2H, I). Histological examination of PC3 xenografted tumors

321

treated with darifenacin showed a reduced percentage of Ki67 positive,

322

proliferating cells and increased percentage of apoptotic cells (Fig. 2J-L). These

323

findings indicate that autocrine activation of CHRM3 promotes prostate cancer

324

growth.

325 326

Autocrine activation of CHRM3 promotes cell migration through regulating

327

epithelial-mesenchymal transition

328

To evaluate the role of CHRM3 in regulating cell migration, we performed

329

transwell assays and scratch tests. In transwell assays, over-expression of

330

CHRM3 in PNT1B cells increased the number of cells that migrated through the

331

membrane (Fig. 3A, B). In scratch tests, over-expression of CHRM3 promoted

332

the confluence of scratched cells (Supplementary Fig. S2A, B). To rule out the

333

potential effects of cell proliferation on cell migration, we normalized the results in

334

transwell assay and scratch test with the total number of cells at the same

335

detection time point as described in the materials and methods section. On the

336

contrary, knock-down of CHRM3 in PC3 cells inhibited their migration capability

337

both in the transwell assay and in the scratch test (Fig. 3C, D and Supplementary

338

Fig. S2C, D). Similarly, the cell migration results were also normalized to the total

339

number of cells at the same detection time point.

340 341

Epithelial-mesenchymal transition (EMT) is an important process in epithelial

342

cancer progression, which facilitates cell migration and invasion (15). To verify

343

whether the activation of CHRM3 could induce EMT, we over-expressed CHRM3

344

in PNT1B cells. We observed that CHRM3 up-regulation caused the PNT1B cells

345

to become less attached to each other and exhibit a mesenchymal phenotype

346

(Fig. 3E). Immunostaining of these cells with E-cadherin, N-cadherin and

13 Downloaded from clincancerres.aacrjournals.org on July 23, 2015. © 2015 American Association for Cancer Research.

Author Manuscript Published OnlineFirst on June 12, 2015; DOI: 10.1158/1078-0432.CCR-14-3163 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

347

Vimentin antibodies confirmed that E-cadherin expression was decreased in

348

CHRM3 over-expressing PNT1B cells; N-cadherin and Vimentin expression was

349

sharply increased (Fig. 3E). Western blotting analysis confirmed the decreased

350

E-cadherin protein level and increased Vimentin protein level in CHRM3 over-

351

expressing PNT1B cells (Fig. 3F). Consistently, real-time PCR analysis also

352

showed an increase in mesenchyme-related gene expression, while E-cadherin

353

was down-regulated in these CHRM3 over-expressing PNT1B cells (Fig. 3G). On

354

the contrary, CHRM3 knock-down PC3 cells reversed its mesenchymal status

355

and expressed more E-cadherin and less N-cadherin (Fig. 3H). The

356

immunoblotting analysis also confirmed that CHRM3 knock-down reversed EMT

357

in PC3 cells (Fig. 3I). In addition, real-time PCR analysis of several EMT-related

358

genes also showed that CHRM3-silenced PC3 cells underwent an opposite

359

process of EMT, that is, mesenchymal-epithelial transition (Fig. 3J). These

360

findings together indicate that autocrine activation of CHRM3 promotes prostate

361

cell migration by regulating EMT.

362 363

CHRM3 is up-regulated in castration-resistant prostate cancer cells

364

To evaluate the role of CHRM3 in castration resistance, a very important feature

365

of prostate cancer, we first measured CHRM3 expression in paired CRPC and

366

androgen-dependent prostate cancer cells: C4-2B and LNCaP, PC3 and PC3-

367

AR+. C4-2B was a bone metastatic and castration-resistant subline of androgen-

368

dependent LNCaP cells (16). PC3-AR+ cells were generated through stable

369

expression of full-length human androgen receptor (AR) in PC3 cells (17). Re-

370

expression of AR restored the response to androgens in PC3-AR+ cells (17,18).

371

Real-time PCR analysis revealed higher CHRM3 mRNA levels in castration-

372

resistant C4-2B and PC3 cells than their paired androgen-dependent LNCaP and

373

PC3-AR+ cells, respectively (Fig. 4A). Secondly, we treated LNCaP cells with

374

bicalutamide, a clinically-used androgen deprivation agent, to imitate the

375

androgen deprivation condition in vitro. After several generations, the expression

376

of CHRM3 was obviously up-regulated in bicalutamide treated LNCaP cells 14 Downloaded from clincancerres.aacrjournals.org on July 23, 2015. © 2015 American Association for Cancer Research.

Author Manuscript Published OnlineFirst on June 12, 2015; DOI: 10.1158/1078-0432.CCR-14-3163 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

377

(Supplementary Fig. S3A). Thirdly, we subcutaneously implanted PC3-AR+ cells

378

in nude mice. When tumors grew for approximately 1 month, we castrated the

379

recipient mice. We analyzed CHRM3 mRNA levels before and post castration.

380

We found that the expression of CHRM3 was significantly increased after

381

castration (Fig. 4B). These findings are in agreement with the GEO database

382

indicating that androgen deprivation resulted in enhanced CHRM3 expression in

383

LuCaP35 cells (Supplementary Fig. S3B) (19). All of these data together suggest

384

a positive correlation of CHRM3 with castration resistance.

385 386

Over-expression of CHRM3 promotes castration resistant growth

387

To evaluate whether activation of CHRM3 could cause castration resistance in

388

vivo, we established tumor xenografts in castrated nude mice. At first, we wanted

389

to establish in vivo tumor models with LNCaP cells. However, LNCaP cells failed

390

to efficiently and consistently form tumors in normal, un-castrated nude mice

391

even with ten million cells. Then, we engrafted PC3-AR+ and PC3 cells

392

subcutaneously in castrated nude mice. We observed that PC3 cells transfected

393

with control lentivirus indeed formed tumors in androgen deprivation conditions

394

(Fig. 4C). On the contrary, PC3-AR+ cells transfected with control lentivirus failed

395

to do so under the same conditions (Fig. 4C). More importantly, over-expression

396

of CHRM3 in these PC3-AR+ cells caused them to re-gain the castration

397

resistant capability and form tumors under conditions of hormone deprivation

398

condition (Fig. 4C). Consistently, immunofluorescent staining of Ki67 and cleaved

399

caspase-3 in tumor sections showed increased cell proliferation and decreased

400

cell apoptosis, respectively, in CHRM3 over-expressed xenografts (Fig. 4D-G).

401

These results suggest that activation of CHRM3 can enhance castration-resistant

402

growth capability of the androgen-dependent prostate cancer cells.

403 404

Blockade of CHRM3 inhibits the castration resistant growth of PC3-luc cells

15 Downloaded from clincancerres.aacrjournals.org on July 23, 2015. © 2015 American Association for Cancer Research.

Author Manuscript Published OnlineFirst on June 12, 2015; DOI: 10.1158/1078-0432.CCR-14-3163 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

405

To study whether the blockade of CHRM3 could influence castration-resistant

406

growth or the sensitivity of prostate cancer cells under androgen deprivation

407

conditions, we established an orthotopic prostate cancer model with luciferase

408

stably expressing PC3 (PC3-luc) cells under an androgen deprivation paradigm,

409

which is more similar to the in situ castration resistant prostate cancer. Seven

410

weeks after implantation, we examined tumor growth by bioluminescence (Fig.

411

5A). Stable silencing of CHRM3 reduced tumor growth when compared with the

412

scrambled shRNA transfected group (Fig. 5B, C). When these primary

413

xenografted tumor sections were processed for immunofluorescent staining with

414

anti-Ki67 antibody to detect the proliferating cells, we observed decreased Ki67

415

positive nuclei in CHRM3 knock-down tumors (Fig. 5D, E). Similarly, when the

416

tumor recipient mice were treated with vehicle or darifenacin at a dosage of 1

417

mg/kg/day, darifenacin significantly inhibited castration resistant growth of PC3-

418

luc cells when compared to the vehicle group (Fig. 5F-H). In addition, darifenacin

419

improved the survival status in tumor-bearing recipient mice (Fig. 5I).

420

Immunostaining of Ki67 also showed decreased cell proliferation in darifenacin

421

treated tumors (Fig. 5J). These findings together confirm that blockade of

422

CHRM3 enhances the sensitivity of androgen-independent PC3-luc cells to

423

androgen deprivation.

424 425

Activation of CHRM3 promotes CaM/CaMKK-dependent phosphorylation of

426

Akt.

427

Next, we wanted to explore the mechanism of autocrine cholinergic signaling in

428

regulating prostate cancer growth and castration resistance. Previous studies

429

reported that Ca2+ influx could promote the phosphorylation of Akt (20-22).

430

Because our data had shown that activation of muscarinic receptors stimulated

431

Ca2+ influx in prostate cancer cells, we next wanted to determine whether the

432

autocrine activation of cholinergic signaling could enhance calcium signaling-

433

mediated phosphorylation of Akt. As shown in Fig. 6A, over-expression of

434

CHRM3 in LNCaP cells increased the phosphorylation of Akt due to the 16 Downloaded from clincancerres.aacrjournals.org on July 23, 2015. © 2015 American Association for Cancer Research.

Author Manuscript Published OnlineFirst on June 12, 2015; DOI: 10.1158/1078-0432.CCR-14-3163 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

435

production of endogenous acetylcholine from the LNCaP cells (Fig. 6A). On the

436

contrary, silencing of CHRM3 in PC3 cells decreased the phosphorylation of Akt

437

(Fig. 6B). These findings indicate that the autocrine cholinergic signaling could

438

promote Akt phosphorylation.

439

To confirm that Akt phosphorylation induced by the autocrine cholinergic

440

signaling is calcium-signaling dependent, we first treated PC3 cells with the

441

CHRM3-specific antagonist darifenacin and the calmodulin-selective antagonist

442

W-7. Western blotting analysis revealed that both darifenacin and W-7 could

443

effectively inhibit Akt phosphorylation stimulated by carbachol (Fig. 6C). Next, we

444

treated PC3 cells with CaMKK antagonist STO-609 to determine whether the

445

downstream signaling of CaM was involved in Akt phosphorylation. STO-609

446

could also effectively block Akt phosphorylation induced by carbachol (Fig. 6D).

447

Furthermore, co-immunoprecipitation showed a direct binding of Akt to CaMKKα

448

(Fig. 6E). These data suggest that autocrine cholinergic signaling promotes

449

prostate cancer growth and castration resistance through CaM/CaMKK-mediated

450

activation of Akt (Fig. 6F).

451 452

Discussion

453

Although a previous study reported a role for neuronal cholinergic signaling in

454

prostate cancer metastasis (3), the present study demonstrates several different

455

and novel findings. First, we found the autonomous expression of ChAT and

456

synthesis of acetylcholine in prostate epithelial cells, suggesting the presence of

457

autocrine cholinergic signaling in the prostate epithelium. Second, we detected

458

an up-regulation of CHRM3 in human prostate cancer tissues compared to their

459

adjacent non-cancer tissues, implicating that CHRM3 might be an additional

460

diagnostic marker of prostate cancer. Third, different from the parasympathetic

461

cholinergic signaling that regulated the microenvironment to promote prostate

462

cancer metastasis (3), we found that direct over-expression or knock-down of

463

CHRM3 in the prostate cells significantly promoted or inhibited cell migration

17 Downloaded from clincancerres.aacrjournals.org on July 23, 2015. © 2015 American Association for Cancer Research.

Author Manuscript Published OnlineFirst on June 12, 2015; DOI: 10.1158/1078-0432.CCR-14-3163 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

464

through the regulation of EMT. Finally, we found that activation or inhibition of

465

CHRM3 promoted or inhibited prostate cancer growth and castration resistance

466

both in vitro and in vivo. Thus, our data strongly indicate that there is an

467

autocrine activation of CHRM3 in prostate cancer epithelial cells.

468 469

Our study shows, for the first time, the secretion of autocrine acetylcholine from

470

prostate epithelial cells and cancer cells. Immunostaining indicated the

471

expression of ChAT in both mouse and human prostate epithelia. In addition, we

472

detected the production of acetylcholine by human prostate epithelial cells,

473

including both cancer cells and non-cancer cells. The concentration of autocrine

474

acetylcholine in the cell culture medium is approximately 2~4 μM, which is

475

sufficient to stimulate Ca2+ influx that can be induced by 0.1 μM of acetylcholine

476

or carbachol (Supplementary Fig. S4). Considering that in the microenvironment

477

of prostate cancer, the concentration of acetylcholine in prostate cancer tissues

478

may be even higher than the acetylcholine secreted into the medium due to

479

higher cell density, such autocrine acetylcholine signaling likely functions in the

480

prostate in vivo. Further support for our autocrine cholinergic signaling model

481

also comes from the results reported previously in other tissue adenocarcinoma

482

cells, such as colon cancer cells and small cell lung cancer cells (7,8).

483 484

The present study provides important insights into the mechanism for the

485

biological effects of activation of muscarinic receptors. First, either the CHRM3-

486

specific inhibitor darifenacin or the calmodulin-selective antagonist W-7 can

487

effectively inhibit Akt phosphorylation stimulated by carbachol. Second, CaMKK

488

antagonist STO-609, which is a downstream signaling component of CaM, also

489

effectively blocks Akt phosphorylation induced by activation of CHRM3. Third,

490

there is direct binding between Akt and CaMKK based on co-immunoprecipitation

491

assays. Fourth, calcium signaling has been shown to mediate Ca2+ influx-

492

induced Akt phosphorylation (22-24). Finally, a blockade of Akt activity has been

18 Downloaded from clincancerres.aacrjournals.org on July 23, 2015. © 2015 American Association for Cancer Research.

Author Manuscript Published OnlineFirst on June 12, 2015; DOI: 10.1158/1078-0432.CCR-14-3163 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

493

shown to suppress castration-resistant growth in both mouse models and clinical

494

settings (25-28). These findings together suggest that autocrine cholinergic

495

signaling promotes prostate cancer growth and castration resistance through the

496

CaM/CaMKK-mediated activation of Akt (Fig. 6F).

497 498

Castration resistance is a major challenge in prostate cancer treatment. However,

499

an effective approach for targeting castration resistant prostate cancer is

500

currently unavailable. In this study, we found that CHRM3 was upregulated in

501

CRPC cells compared to matched androgen-dependent cells. While over-

502

expression of CHRM3 was sufficient to cause androgen-dependent PC3-AR+

503

cells to form tumors in castrated mice, stable silencing of CHRM3 inhibited the

504

castration resistant growth of PC3 cells in orthotopic xenografts. Notably, our

505

study showed that the CHRM3-specific antagonist darifenacin was effective to

506

inhibit PC3 cell growth in castrated nude mice. Given that specific antagonists of

507

CHRM3 have been widely used in clinical conditions such as OAB (overactive

508

bladder) and COPD (chronic obstructive pulmonary diseases), clinical trials with

509

such CHRM3 antagonists are warranted and may hold promise for the treatment

510

of primary prostate cancer as well as castration-resistant prostate cancer.

511 512 513 514 515 516 517 518

19 Downloaded from clincancerres.aacrjournals.org on July 23, 2015. © 2015 American Association for Cancer Research.

Author Manuscript Published OnlineFirst on June 12, 2015; DOI: 10.1158/1078-0432.CCR-14-3163 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

519

References

520 521 522 523 524 525 526 527 528 529 530 531 532 533 534 535 536 537 538 539 540 541 542 543 544 545 546 547 548 549 550 551 552 553 554 555 556 557 558 559 560 561 562 563 564

1. 2. 3. 4. 5. 6.

7.

8.

9.

10. 11.

12.

13.

14. 15. 16.

17.

Siegel R, Ma J, Zou Z, Jemal A. Cancer statistics, 2014. CA: a cancer journal for clinicians 2014;64(1):9-29. Shen MM, Abate-Shen C. Molecular genetics of prostate cancer: new prospects for old challenges. Gene Dev 2010;24(18):1967-2000. Magnon C, Hall SJ, Lin J, Xue X, Gerber L, Freedland SJ, et al. Autonomic nerve development contributes to prostate cancer progression. Science 2013;341(6142). Wessler IK, Kirkpatrick CJ. Activation of muscarinic receptors by non-neuronal acetylcholine. Muscarinic Receptors: Springer; 2012. p 469-91. Spindel ER. Muscarinic receptor agonists and antagonists: effects on cancer. Muscarinic Receptors: Springer; 2012. p 451-68. Gutkind JS, Novotny EA, Brann MR, Robbins KC. Muscarinic acetylcholine receptor subtypes as agonist-dependent oncogenes. Proceedings of the National Academy of Sciences 1991;88(11):4703-07. Song P, Sekhon HS, Jia Y, Keller JA, Blusztajn JK, Mark GP, et al. Acetylcholine is synthesized by and acts as an autocrine growth factor for small cell lung carcinoma. Cancer research 2003;63(1):214-21. Cheng K, Samimi R, Xie G, Shant J, Drachenberg C, Wade M, et al. Acetylcholine release by human colon cancer cells mediates autocrine stimulation of cell proliferation. American Journal of Physiology-Gastrointestinal and Liver Physiology 2008;295(3):G591G97. Rayford W, Noble M, Austenfeld M, Weigel J, Mebust W, Shah G. Muscarinic cholinergic receptors promote growth of human prostate cancer cells. The prostate 1997;30(3):16066. Tian L, Fang YX, Xue JL, Chen JZ. Four microRNAs promote prostate cell proliferation with regulation of PTEN and its downstream signals in vitro. PLoS One 2013;8(9):e75885. Hayward SW, Dahiya R, Cunha GR, Bartek J, Deshpande N, Narayan P. Establishment and characterization of an immortalized but non-transformed human prostate epithelial cell line: BPH-1. In vitro cellular & developmental biology Animal 1995;31(1):14-24. Wang J, Ying G, Wang J, Jung Y, Lu J, Zhu J, et al. Characterization of phosphoglycerate kinase-1 expression of stromal cells derived from tumor microenvironment in prostate cancer progression. Cancer research 2010;70(2):471-80. Zheng J, Wang J, Sun X, Hao M, Ding T, Xiong D, et al. HIC1 modulates prostate cancer progression by epigenetic modification. Clinical cancer research : an official journal of the American Association for Cancer Research 2013;19(6):1400-10. Leong KG, Wang BE, Johnson L, Gao WQ. Generation of a prostate from a single adult stem cell. Nature 2008;456(7223):804-8. Yang J, Weinberg RA. Epithelial-mesenchymal transition: At the crossroads of development and tumor metastasis. Dev Cell 2008;14(6):818-29. Thalmann GN, Anezinis PE, Chang SM, Zhau HE, Kim EE, Hopwood VL, et al. Androgenindependent cancer progression and bone metastasis in the LNCaP model of human prostate cancer. Cancer research 1994;54(10):2577-81. Yuan S, Trachtenberg J, Mills GB, Brown TJ, Xu F, Keating A. Androgen-induced inhibition of cell proliferation in an androgen-insensitive prostate cancer cell line (PC-3) transfected with a human androgen receptor complementary DNA. Cancer research 1993;53(6):1304-11.

20 Downloaded from clincancerres.aacrjournals.org on July 23, 2015. © 2015 American Association for Cancer Research.

Author Manuscript Published OnlineFirst on June 12, 2015; DOI: 10.1158/1078-0432.CCR-14-3163 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

565 566 567 568 569 570 571 572 573 574 575 576 577 578 579 580 581 582 583 584 585 586 587 588 589 590 591 592 593 594 595 596

18.

19.

20.

21. 22.

23. 24. 25.

26.

27.

28.

Heisler LE, Evangelou A, Lew AM, Trachtenberg J, Elsholtz HP, Brown TJ. Androgendependent cell cycle arrest and apoptotic death in PC-3 prostatic cell cultures expressing a full-length human androgen receptor. Molecular and cellular endocrinology 1997;126(1):59-73. Sun Y, Wang BE, Leong KG, Yue P, Li L, Jhunjhunwala S, et al. Androgen deprivation causes epithelial-mesenchymal transition in the prostate: implications for androgendeprivation therapy. Cancer research 2012;72(2):527-36. Deb TB, Coticchia CM, Dickson RB. Calmodulin-mediated activation of Akt regulates survival of c-Myc-overexpressing mouse mammary carcinoma cells. Journal of Biological Chemistry 2004;279(37):38903-11. Yano S, Tokumitsu H, Soderling TR. Calcium promotes cell survival through CaM-K kinase activation of the protein-kinase-B pathway. Nature 1998;396(6711):584-87. Coticchia CM, Revankar CM, Deb TB, Dickson RB, Johnson MD. Calmodulin modulates Akt activity in human breast cancer cell lines. Breast cancer research and treatment 2009;115(3):545-60. Yano S, Tokumitsu H, Sodeling TR. Calcium promotes cell survival through CaM-K kinase activation of the protein-kinase-B pathway. Nature 1998;396(6711):584-87. Schmitt JM, Smith S, Hart B, Fletcher L. CaM kinase control of AKT and LNCaP cell survival. J Cell Biochem 2012;113(5):1514-26. Kinkade CW, Castillo-Martin M, Puzio-Kuter A, Yan J, Foster TH, Gao H, et al. Targeting AKT/mTOR and ERK MAPK signaling inhibits hormone-refractory prostate cancer in a preclinical mouse model. The Journal of clinical investigation 2008;118(9):3051. Mulholland DJ, Tran LM, Li Y, Cai H, Morim A, Wang S, et al. Cell autonomous role of PTEN in regulating castration-resistant prostate cancer growth. Cancer cell 2011;19(6):792-804. Gao H, Ouyang X, Banach-Petrosky WA, Gerald WL, Shen MM, Abate-Shen C. Combinatorial activities of Akt and B-Raf/Erk signaling in a mouse model of androgenindependent prostate cancer. Proceedings of the National Academy of Sciences 2006;103(39):14477-82. Floc'h N, Kinkade CW, Kobayashi T, Aytes A, Lefebvre C, Mitrofanova A, et al. Dual targeting of the Akt/mTOR signaling pathway inhibits castration-resistant prostate cancer in a genetically engineered mouse model. Cancer research 2012;72(17):4483-93.

597 598 599 600 601 602 603

21 Downloaded from clincancerres.aacrjournals.org on July 23, 2015. © 2015 American Association for Cancer Research.

Author Manuscript Published OnlineFirst on June 12, 2015; DOI: 10.1158/1078-0432.CCR-14-3163 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

604

Figure legends

605

Figure 1. Presence of a functional autocrine cholinergic system in the prostate

606

epithelium. A, Immunofluorescent staining of Tuj-1 in 2-week-old mouse prostate

607

sections. B, Immunofluorescent images showing a selective epithelial expression

608

pattern of ChAT in the mouse prostate. C, Immunohistochemistry of ChAT in

609

human prostate sections. D, Detection of the release of acetylcholine from

610

various human prostate cell lines. E, Immunohistochemistry of CHRM3 in human

611

prostate cancer tissue arrays. F, Staining intensities of CHRM3 quantified by IOD

612

(Integral optical density); data are analyzed by paired t-test. G, Ca2+ influx

613

detected by Fluo-4 AM in PC3 cells. Acetylcholine (10 μM) and carbachol (10 μM)

614

are used to trigger Ca2+ influx. To counteract Ca2+ influx induced by carbachol,

615

darifenacin (10 μM) is added 5 minutes before the addition of carbachol. H,

616

Detection of Ca2+ influx induced by endogenous acetylcholine. Conditioned

617

medium is collected from PC3 cells cultured in a serum free medium, in which

618

neostigmine (10 μM) was added to inhibit the degradation of acetylcholine. Data

619

are analyzed with Student’s t-test, * P

CaMKK-Mediated Phosphorylation of Akt.

Although a previous study reported nerve ending-derived acetylcholine promoted prostate cancer invasion and metastasis by regulating the microenvironm...
18MB Sizes 1 Downloads 10 Views