monitoring of autophagy is complicated—salinomycin as an example

7
UNCORRECTED PROOF Q1 Monitoring of autophagy is complicatedsalinomycin as an example Q2 Jaganmohan Reddy Jangamreddy a , Soumya Panigrahi b , Marek J. Los a,c, 3 a Department of Clinical and Experimental Medicine (IKE), Division of Cell Biology and Integrative Regenerative Medicine Center (IGEN), Linköping University, Sweden 4 b Department of Medicine/Infectious Diseases, Case Western Reserve University, Cleveland, OH 44106, USA 5 c Department of Pathology, Pomeranian Medical University, Szczecin, Poland abstract 6 article info 7 Article history: 8 Received 4 November 2014 9 Received in revised form 10 December 2014 10 Accepted 11 December 2014 11 Available online xxxx 12 Keywords: 13 Autophagic ux 14 GFP-LC3 15 mTandem GFP-RFP LC3 16 p62/SQSTRM1 17 Vacuolization 18 Salinomycin 19 Monitoring of autophagy is challenging because of its multiple steps and lack of single betting technique for a 20 complete mechanistic understanding, which makes the task complicated. Here, we evaluate the functionality 21 of autophagy triggered by salinomycin (anti-cancer stem cell agent) using ow cytometry and advanced micros- 22 copy. We show that salinomycin does induce functional autophagy at lower concentrations and such a dose is cell 23 type-dependent. For example, PC3 cells show active autophagic ux at 10 μM concentration of salinomycin while 24 murine embryonic broblasts already show an inhibition of ux at such doses. A higher concentration of 25 salinomycin (i.e. 30 μM) inhibits autophagic ux in both cell types. The data conrms our previous ndings 26 that salinomycin is an inducer of autophagy, whereas autophagic ux inhibition is a secondary response. 27 © 2014 Elsevier B.V. All rights reserved. 32 1. Introduction 33 Autophagy is the primary cellular metabolic stress-induced catabolic 34 response that converts organelles, pathogens and cellular debris to its 35 building blocks that are eventually recycled. A dysfunctional autophagy 36 promotes carcinogenesis, while a functional autophagy is necessary for 37 cancer cell survival under extreme conditions of hypoxia and nutrient 38 deprivation [1]. Thus, the denition of chemotherapeutic agents in 39 their role as either inducers or inhibitors of functional autophagy is 40 pivotal for proper planning of treatment. The complexity of autophagic 41 process and the lack of one robust standard technique to follow its pro- 42 gression warrant the employment of several techniques. Standardization 43 of techniques to precisely monitor autophagy is constantly evolving [2]. 44 Macroautophagy, referred to in this article as autophagy, triggered 45 either by activation of class III PI3-kinases or inhibition of class I PI3- 46 kinases, initiates the formation of an initial phagophore that matures 47 to a double-membraned envelope, which engulfs the substrates. The 48 engulfed material is then degraded upon fusion with acidic lysosomes. 49 The initiation of autophagy is a tightly regulated sequential process 50 governed by multiprotein complexes composed of autophagy-related 51 proteins. To name a few examples in sequential order during autophagy 52 initiation: Beclin1, ATG5, ATG7 and LC3 (ATG8) along with other cellu- 53 lar proteins, those that are degraded by autophagy like p62/SQSTRM1 54 are traced to identify functional autophagy. Beclin1 marks the initiation 55 of autophagy, and forms a complex with autophagy initiation multimer 56 involving UVRAG and UMBRA [3]. This initiation is followed by the elon- 57 gation of the phagophore through ATG5- and ATG7-dependent mecha- 58 nisms leading to the lipidation of LC3 to form LC3II and consequent 59 recruitment to the elongating phagophore [3]. Thus, mitigation of 60 LC3II acts as a major tool to evaluate the elongation of the phagophore. 61 LC3II is localized on either side of the membrane. Upon maturation of 62 the autophagosome by fusing with lysosomes, part of LC3II localized 63 to the inner membrane gets degraded while the rest on the outer mem- 64 brane is recycled. Hence, monitoring of LC3 is widely used as it acts not 65 only as a marker for autophagy induction but also to study maturation of 66 the autophagosome. More notably, autophagy monitoring studies using 67 tagged LC3 with either GFP (green orescent protein) alone, or with tan- 68 dem GFP and RFP (red orescent proteins), or most recently, Wasabi as 69 a replacement for GFP has become the standard [4]. Since lysosomes ul- 70 timately dene the active, functional autophagy, staining of these acidic 71 organelles, to determine the number of lysosomes and their localization 72 to autophagosomes (their fusion marks the completion of autophagic 73 process), is also frequently used for monitoring autophagy. 74 Streptomyces albusderived ionophore salinomycin (Sal) is widely 75 studied because of its potency to kill cancer stemlike cells [5,6]. We, 76 along with other groups, have shown that salinomycins toxicity towards 77 cancer and cancer stem cells depends on the disruption of function of 78 mitochondria [6,7]. We employ a wide range of breast cancer, prostate 79 cancer and normal primary human cells to show that salinomycin toxic- 80 ity is concentration-, time- and cell typedependent with least toxicity 81 towards normal primary broblasts [6]. However, there are conicting Biochimica et Biophysica Acta xxx (2014) xxxxxx Corresponding author at: Department of Clinical and Experimental Medicine, Integrative Regenerative Medicine Center (IGEN), Linköping University, Cell Biology Building, Level 10, 581 85 Linköping, Sweden. Tel.: +46 10 10 32787. E-mail address: [email protected] (M.J. Los). BBAMCR-17453; No. of pages: 7; 4C: 2, 3, 4, 5 http://dx.doi.org/10.1016/j.bbamcr.2014.12.022 0167-4889/© 2014 Elsevier B.V. All rights reserved. Contents lists available at ScienceDirect Biochimica et Biophysica Acta journal homepage: www.elsevier.com/locate/bbamcr Please cite this article as: J.R. Jangamreddy, et al., Monitoring of autophagy is complicatedsalinomycin as an example, Biochim. Biophys. Acta (2014), http://dx.doi.org/10.1016/j.bbamcr.2014.12.022

Upload: liu-se

Post on 01-Dec-2023

0 views

Category:

Documents


0 download

TRANSCRIPT

1Q1

2Q2

345

6

7891011

12131415161718

32

33

34

35

36

37

38

39

40

41

42

43

44

45

46

47

48

49

50

51

52

53

Biochimica et Biophysica Acta xxx (2014) xxx–xxx

BBAMCR-17453; No. of pages: 7; 4C: 2, 3, 4, 5

Contents lists available at ScienceDirect

Biochimica et Biophysica Acta

j ourna l homepage: www.e lsev ie r .com/ locate /bbamcr

Monitoring of autophagy is complicated—salinomycin as an example

F

Jaganmohan Reddy Jangamreddy a, Soumya Panigrahi b, Marek J. Łos a,c,⁎a Department of Clinical and Experimental Medicine (IKE), Division of Cell Biology and Integrative Regenerative Medicine Center (IGEN), Linköping University, Swedenb Department of Medicine/Infectious Diseases, Case Western Reserve University, Cleveland, OH 44106, USAc Department of Pathology, Pomeranian Medical University, Szczecin, Poland

O

⁎ Corresponding author at: Department of ClinicalIntegrative Regenerative Medicine Center (IGEN), LinkBuilding, Level 10, 581 85 Linköping, Sweden. Tel.: +46 1

E-mail address: [email protected] (M.J. Łos).

http://dx.doi.org/10.1016/j.bbamcr.2014.12.0220167-4889/© 2014 Elsevier B.V. All rights reserved.

Please cite this article as: J.R. Jangamreddy, e(2014), http://dx.doi.org/10.1016/j.bbamcr.2

Oa b s t r a c t

a r t i c l e i n f o

19

20

21

22

23

24

25

26

27

Article history:Received 4 November 2014Received in revised form 10 December 2014Accepted 11 December 2014Available online xxxx

Keywords:Autophagic fluxGFP-LC3mTandem GFP-RFP LC3p62/SQSTRM1VacuolizationSalinomycin

D P

R

Monitoring of autophagy is challenging because of its multiple steps and lack of single befitting technique for acomplete mechanistic understanding, which makes the task complicated. Here, we evaluate the functionalityof autophagy triggered by salinomycin (anti-cancer stem cell agent) using flow cytometry and advancedmicros-copy.We show that salinomycin does induce functional autophagy at lower concentrations and such a dose is celltype-dependent. For example, PC3 cells show active autophagic flux at 10 μMconcentration of salinomycinwhilemurine embryonic fibroblasts already show an inhibition of flux at such doses. A higher concentration ofsalinomycin (i.e. 30 μM) inhibits autophagic flux in both cell types. The data confirms our previous findingsthat salinomycin is an inducer of autophagy, whereas autophagic flux inhibition is a secondary response.

© 2014 Elsevier B.V. All rights reserved.

E

T

54

55

56

57

58

59

60

61

62

63

64

65

66

67

68

69

70

71

72

73

74

75

76

UNCO

RREC1. Introduction

Autophagy is the primary cellularmetabolic stress-induced catabolicresponse that converts organelles, pathogens and cellular debris to itsbuilding blocks that are eventually recycled. A dysfunctional autophagypromotes carcinogenesis, while a functional autophagy is necessary forcancer cell survival under extreme conditions of hypoxia and nutrientdeprivation [1]. Thus, the definition of chemotherapeutic agents intheir role as either inducers or inhibitors of functional autophagy ispivotal for proper planning of treatment. The complexity of autophagicprocess and the lack of one robust standard technique to follow its pro-gressionwarrant the employment of several techniques. Standardizationof techniques to precisely monitor autophagy is constantly evolving [2].

Macroautophagy, referred to in this article as autophagy, triggeredeither by activation of class III PI3-kinases or inhibition of class I PI3-kinases, initiates the formation of an initial phagophore that maturesto a double-membraned envelope, which engulfs the substrates. Theengulfed material is then degraded upon fusion with acidic lysosomes.The initiation of autophagy is a tightly regulated sequential processgoverned by multiprotein complexes composed of autophagy-relatedproteins. To name a few examples in sequential order during autophagyinitiation: Beclin1, ATG5, ATG7 and LC3 (ATG8) along with other cellu-lar proteins, those that are degraded by autophagy like p62/SQSTRM1

77

78

79

80

81

and Experimental Medicine,öping University, Cell Biology0 10 32787.

t al., Monitoring of autophag014.12.022

are traced to identify functional autophagy. Beclin1marks the initiationof autophagy, and forms a complex with autophagy initiation multimerinvolvingUVRAGandUMBRA [3]. This initiation is followed by the elon-gation of the phagophore through ATG5- and ATG7-dependent mecha-nisms leading to the lipidation of LC3 to form LC3II and consequentrecruitment to the elongating phagophore [3]. Thus, mitigation ofLC3II acts as a major tool to evaluate the elongation of the phagophore.LC3II is localized on either side of the membrane. Upon maturation ofthe autophagosome by fusing with lysosomes, part of LC3II localizedto the inner membrane gets degradedwhile the rest on the outer mem-brane is recycled. Hence, monitoring of LC3 is widely used as it acts notonly as amarker for autophagy induction but also to studymaturation ofthe autophagosome. More notably, autophagy monitoring studies usingtagged LC3with either GFP (greenflorescent protein) alone, orwith tan-dem GFP and RFP (red florescent proteins), or most recently, Wasabi asa replacement for GFP has become the standard [4]. Since lysosomes ul-timately define the active, functional autophagy, staining of these acidicorganelles, to determine the number of lysosomes and their localizationto autophagosomes (their fusion marks the completion of autophagicprocess), is also frequently used for monitoring autophagy.

Streptomyces albus–derived ionophore salinomycin (Sal) is widelystudied because of its potency to kill cancer stem–like cells [5,6]. We,alongwith other groups, have shown that salinomycin’s toxicity towardscancer and cancer stem cells depends on the disruption of function ofmitochondria [6,7]. We employ a wide range of breast cancer, prostatecancer and normal primary human cells to show that salinomycin toxic-ity is concentration-, time- and cell type–dependent with least toxicitytowards normal primary fibroblasts [6]. However, there are conflicting

y is complicated—salinomycin as an example, Biochim. Biophys. Acta

82

83

84

85

86

87

88

89

90

91

92

93

94

95

96

97

98

99

100

101

102

103

104

105

106

107

108

109

110

111

112

113

114

115

116

117

118

119

120

121

122

123

124

125

126

2 J.R. Jangamreddy et al. / Biochimica et Biophysica Acta xxx (2014) xxx–xxx

reports regarding autophagy induction by salinomycin. We, along withLi and colleagues [8], show that salinomycin induces autophagic flux,while Yue and collaborators [9] show salinomycin as an inhibitor of func-tional autophagy without altering the lysosomal acidity but throughan unknown mechanism of attenuating lysosomal proteases. The com-parison is complicated by the fact that all the above-mentioned studiesemployed different cancer cellular models [6,8,9]. So, in this brief report,we evaluate autophagy induced by salinomycin employing a variety oftechniques including recently developed flow cytometry techniques,and live cell imaging approaches [10–12]. We show that salinomycin-induced autophagic flux is dose-, and cell type–dependent.

2. Results

2.1. Salinomycin is an inducer of autophagy and autophagic flux

We initially identified the salinomycin function in regulating autoph-agic flux using normal cells (MEF) and cancer cells (PC3 and SKBR3). Asa follow-up to our previous study, we assessed by Western blot theratiometric increase of LC3II accumulation in the presence of salinomycinand chloroquine compared to either salinomycin or chlorquine alone [6].As shown in Fig. 1A and Supplementary Fig. 1A, salinomycin-treated cells(0.1 μM and 1 μM) and chloroquine-treated cells (25 μM) showedincreased accumulation of LC3II, but only cells treated with 0.1 μM Salshowed a ratiometric increase in the presence of chloroquine. Even

UNCO

RRECT

A

B

Fig. 1. Salinomycin induces autophagic flux. Autophagic fluxwasmonitored through estimatingtions. LC3II showed increased levels upon salinomycin treatment (0.1 μM and 1 μM), but in the(A). Even though 1 μM salinomycin-treated samples showed increase in LC3II accumulation indegrade upon autophagy induction by salinomycin treatment, showing a functional autophagyimages of MEF cells expressing GFP-LC3 and treated with salinomycin show increased LC3 puntreated cells show huge vacuoles with GFP-LC3 and clumping of vacuoles near the nucleus (B)

Please cite this article as: J.R. Jangamreddy, et al., Monitoring of autophag(2014), http://dx.doi.org/10.1016/j.bbamcr.2014.12.022

RO

OF

though 1 μMSal (Fig. 1A) and 10 μMSal (Supplementary Fig. 1B) treatedcells showed increased accumulation of LC3II in the presence of chloro-quine, they did not show a significant difference compared to 1 μM Saland 10 μMSal treated cells alone, respectively. To remedy the possibilitythat inhibition of autophagy by 25 μM Cq would only be partial, we in-creased the concentration of chloroquine to 50 μM. In the presence of50 μM Cq, a further increase in accumulation of LC3II occurred upon10 μM Sal treatment as compared to Cq and 10 μM Sal-treated samplesindependently (Supplementary Fig. 1B).

Autophagic flux was further monitored using another autophagicmarker, p62/SQSTRM1, and the results correlated with that of LC3IIflux. In the presence of salinomycin at 0.1 and 1 μM concentration,p62/SQSTRM1 was directed towards proteolysis in PC3 and MEFcells. The proteolysis of p62/SQSTRM1 was resistant to Cq pretreated,inferring that salinomycin does induce functional autophagy at lowerconcentrations (Fig. 1A and Supplementary Fig. 1A). However, therewas no increase of LC3 flux in SKBR3 cells, and p62/SQSTRM1 levelsremained unaltered at 1 μM Sal regardless of the presence of chloro-quine (Supplementary Fig. 1A). Thus, the dosage of salinomycin differ-entially affects autophagic flux among cell types.

2.2. Higher doses of salinomycin inhibits autophagic flux

Based on our observations so far we hypothesized that salinomycincould inhibit autophagic flux at higher concentrations depending on

ED P

LC3II and p62/SQSTM1 protein levels upon salinomycin treatment at different concentra-presence of chloroquine 0.1 μM salinomycin-treated samples showed ratiometric increasethe presence of chloroquine it was only a linear increase (A). p62/SQSTM1 protein levelsand this degradation was inhibited upon chloroquine treatment (A). Confocal microscopyctae (B) but 1 μM salinomycin-treated cells show distinct dots (punctae) while the 30 μM. N = 3, *p b 0.05.

y is complicated—salinomycin as an example, Biochim. Biophys. Acta

127

128

129

130

131

132

133

134

135

136

137

138

3J.R. Jangamreddy et al. / Biochimica et Biophysica Acta xxx (2014) xxx–xxx

cell type as different cells have different toxicity doses of salinomycin.To test this, a follow-up study with 1 μM and 30 μM salinomycin wasdone. As shown in Fig. 1B, GFP-LC3 expressing MEF cells treated with1 μM salinomycin showed distinct green autophagic punctae while thecontrol cells show homogeneous green florescence throughout thecytoplasm. Even though cells treated with higher concentrations of

UNCO

RRECT

A

B

Fig. 2. Total or membrane-bound GFP-LC3 quantification for autophagic flux determination. GFinhibitor chloroquine or alone are subjected to flow cytometry to estimate the total green flordecreased median florescence intensity (MFI) of green florescence compared to control. Chlothe presence of autophagic inducer (salinomycin or rapamycin) or by itself. Samples treatedthat salinomycin at higher concentrations inhibits autophagic flux (upper panel: A). MEF cellsestimation of LC3 bound to autophagic vacuoles shows increase in MFI both in the presenceautophagy (1 μM Sal and 0.25 μM Rap). Chloroquine pretreated cells in the presence of 1 μM SSal (lower panel: A). Quantitative representation of MFI values either in the presence or absen

Please cite this article as: J.R. Jangamreddy, et al., Monitoring of autophag(2014), http://dx.doi.org/10.1016/j.bbamcr.2014.12.022

salinomycin showed green punctae, many cells exhibit heterogeneityin the punctae pattern as some cells show appearance of clumped GFPvacuoles while others show distinct autophagic punctae (Fig. 1B).

Following this initial observation of distinct LC3II pattern of accumu-lation among cells treated with different concentrations of salinomycin,we employedflowcytometric approaches to enumerate the salinomycin

ED P

RO

OF

P-LC3 expressing MEF cells treated with salinomycin either in the presence of autophagyescence. Treatment with 1 μM salinomycin or 0.25 μM rapamycin (positive control) showroquine treatment shows an increase in MFI due to the inhibition of autophagy either inwith 30 μM of salinomycin show similar increase in MFI as chloroquine and thus showwere treated with saponin to remove the cytoplasmic unbound GFP-LC3 to facilitate theof inhibitors of functional autophagy (chloroquine and 30 μM Sal) as well as inducers ofal and 0.25 μM Rap shows tremendous increase in MFI but not in the presence of 30 μMce of saponin for the above (B). N = 4, *p b 0.05.

y is complicated—salinomycin as an example, Biochim. Biophys. Acta

139

140

141

142

143

144

145

146

147

148

149

150

151

152

153

154

155

156

157

158

159

160

161

162

163

164

165

166

167

168

169

170

4 J.R. Jangamreddy et al. / Biochimica et Biophysica Acta xxx (2014) xxx–xxx

triggered autophagic flux using GFP-LC3 expressing cells as describedpreviously by Shvets et al. and Eng et al. [10–12]. In the presence ofactive autophagy, GFP-LC3 that is located to the inner membraneof autophagosome gets degraded, while the rest is recycled a partialdecrease in green florescence is observed among cells treated with1 μM Sal and 0.25 μM rapamycin (positive control). However, 30 μMSal, similar to 50 μM Cq, shows increased accumulation (shift to theright in relevant histograms; Fig. 2A, upper panel). This shows thatsalinomycin at higher concentrations inhibits autophagic flux, whileat lower concentrations, it is an inducer of functional autophagy. Greenflorescence even though showed partial increase in the presence ofboth salinomycin and Cq, it did not show a ratiometric increase.

Next, we quantified the amount of GFP-LC3 localized to theautophagosome membrane. To do that, we removed free, cytoplasmicGFP-LC3 by permeabilization, thus we were able to directly quantifythe autophagosome florescence [12]. As shown in Fig. 2A (lower

UNCO

RRECT

A

B

Fig. 3. The dose of salinomycin that inhibits autophagic flux depends on cell type. MEF Cells ovflorescence better than the red florescence while 30 μM Sal and chloroquine show an increasethe same pattern as MEF cells but even at 10 μM Sal shows active autophagic flux with decreas(B). N = 3.

Please cite this article as: J.R. Jangamreddy, et al., Monitoring of autophag(2014), http://dx.doi.org/10.1016/j.bbamcr.2014.12.022

F

panel), cells permeabilized with 0.05% saponin after treatment witheither inhibitor of autophagy or inducers, showed an increase of greenflorescence similar to the observation made using Western blotting.As expected, salinomycin (1 μM and 30 μM), rapamycin (1 μM) andCq (50 μM) caused the increase of green florescence due to a rise inautophagosome quantity. Similar to Western blotting results, 1 μMSal in the presence of Cq showed a ratiometric increase in greenflorescence compared to 1 μM Sal or Cq alone; however, 30 μM Saldid not show such increase. This data indicates that salinomycin athigher concentrations does inhibit autophagic flux. Similarly, flowcytometric quantification of green and red fluorescence among cellsexpressing mTandem GFP-RFP LC3 showed lower green and redflorescence in the presence of autophagic inducers (1 μM Sal, 1 μMrapamycin, and starvation), while chloroquine and 30 μM Sal showeda decrease in both green and red florescence in both MEF and PC3cells (Fig. 3AB).

ED P

RO

O

erexpressing mTandem-RPF-GFP-LC3 treated with 1 μM Sal for 6 h show decreased greenin green and red florescence compared to control cells (A). Similarly, PC3 cells also showed green and red florescence compared to control as rapamycin and starvation conditions

y is complicated—salinomycin as an example, Biochim. Biophys. Acta

171

172

173

174

175

176

177

178

179

180

181

182

183

184

185

186

187

188

189

190

191

192

193

194

195

196

197

198

5J.R. Jangamreddy et al. / Biochimica et Biophysica Acta xxx (2014) xxx–xxx

2.3. Live cell monitoring of autophagy

Additionally, we monitored autophagic processes using live cellimaging of GFP-LC3 expressing cells stained with Lysotracker redafter treatment with 1 μM Sal for 30 min. As shown in the Fig. 4A,lysosomes are co-localized with autophagosomes by the 10th frame(100 s) and a progressive degradation of green florescence is ob-served with time along with final decrease in lysosome count. Wefurther quantified total number of green punctae and lysosomes pertime point, and observed an increase in the number of lysosomesand green punctae together with very synchronous fluctuations inthe number of lysosomes and autophagosomes (Fig. 4B,C). Thus, effec-tively showing that salinomycin induces active autophagy that func-tions to degrade cellular organelles and proteins to counteract thecellular stress.

UNCO

RRECT

A

B C

Fig. 4. Co-localization of autophagosomes and lysosomes upon salinomycin treatment. GFP-LC3Sal were imaged using time-lapse live cell microscopy with 10 s interval for 1 h. Red-stainedegradation of green and red florescence is observed from frame 100 (A). Using Bitplane Iautophagosomal count is estimated for each time frame. Frame 50 shows an increase in lysto frame 10 (B). Graphical representation of lysosomal and autophagosomal count over the pan increase in autophagosomal count followed by increased lysosomal count and eventual deg

Please cite this article as: J.R. Jangamreddy, et al., Monitoring of autophag(2014), http://dx.doi.org/10.1016/j.bbamcr.2014.12.022

3. Discussion

The discovery of salinomycin’s cancer stem cell targeting ability,in 2009, lead to intensive studies of its molecular mechanism of ac-tion [5]. We, and others, have studied the autophagy triggered bysalinomycin [6,8,9]. Even though salinomycin is well-known to in-duce autophagy that functions as a cell-protective mechanism, theunderlying mechanism is not fully understood, with some conflictingreports recently published. For example, Yue and colleagues, in theirelaborate study using MCF7 and HMLER cancer stem cells, show thatsalinomycin acts as an inhibitor of autophagic flux [9]. On the contrary,in our experimental system involving PC3 (prostate cancer cells), SKBR3(breast cancer cells) and primary human dermal fibroblast cells, as wellas cells used in the original paper by Li et al., we observed an activeautophagic flux in salinomycin-treated cells [6,8].

ED P

RO

OF

overexpressingMEF cells stainedwith lysotracker dye for 30min and treatedwith 0.1 μMd lysosomes co-localize with the GFP-LC3 autophagosomes (frame 30) and an eventualmaris software to analyze 3D images obtained with time-lapse studied lysosomal andosomal (red) count and their co-localization with green autophagic punctae comparederiod of time among salinomycin-treated cells show a similar trend in fluctuations withradation (C). N = 3.

y is complicated—salinomycin as an example, Biochim. Biophys. Acta

T

199

200

201

202

203

204

205

206

207

208

209

210

211

212

213

214

215

216

217

218

219

220

221

222

223

224

225

226

227

228

229

230

231

232

233

234

235

236

237

238

239

240

241

242

243

244

245

246

247

248

249

250

251

252

253

254

255

256

257

258

259

260

261

262

263

264

265

266

267

268

269

270

271

272

273

274

275

276

277

278

279

280

281

282

283

284

285

286

287

288

289

290

291

292

293

294

295

296

297

298

299

300

301

302

303

304

305

306

307

308

309

310

311

312

313

314

315

316

317

318

319

6 J.R. Jangamreddy et al. / Biochimica et Biophysica Acta xxx (2014) xxx–xxx

UNCO

RREC

To understand these contradictory observations, in this study, wefurther examined in detail salinomycin’s role in autophagic flux, usingboth normal murine embryonic fibroblasts and prostate cancer PC3cells. Utilizing the densitometric analysis of LC3II expression in thepresence of salinomycin and/orwith autophagic inhibitors (chloroquineor bafilomycin), our study, as well as those conducted by Li and col-leagues, shows a ratiometric increase at lower concentrations as com-pared to individual treatments. However, only a partial increase ofLC3II was observed among cells treated with higher concentrations ofsalinomycin (Fig. 1A and Supplementary Fig. 1). Furthermore, in thisstudy, the observation that 25 μM concentration of chloroquine treat-ment was only partially able to inhibit LC3II turnover triggered bysalinomycin at higher concentration (10 μM). A concentration of 50 μMchloroquine shows a rise in LC3II accumulation and displays the impor-tance of effectively blocking autophagy activity depending on the ex-tent of the autophagy trigger. Previous studies also show that LC3IIpunctae are increased and co-localizewith lysosomes that remain acidicupon salinomycin treatment [6,9]. However, Yue et al. further show thatsalinomycin inhibits lysosomal proteases thus triggering the inhibitionof autophagic flux [9].

Because of the limitations of microscopy in analyzing minor fluctua-tions,weusedflowcytometric quantification offlorescence. As presentedin Fig. 2 and Supplementary Fig. 3, GFP-LC3 expressing MEF and PC3, re-spectively, treatment with 1 μM salinomycin showed a similar trendas positive control rapamycin in decreasing green florescence intensity(increased autophagy). However, 30 μMsalinomycin shows an increaseof the green florescence (decrease of autophagy) as autophagy inhibitorchloroquine. Similar observations were made with mTandem GFP-RFPLC3 with both green and red florescence (Fig. 3A,B). However, PC3cells show decreased green and red florescence even at 10 μM concen-tration (Fig. 3B). This demonstrates that salinomycin-mediated inhibi-tion of autophagic flux is dependent on cell type and dosage.

While conducting studies involving GFP-LC3 overexpression invarious cell lines, we observed that very high expression levels of theGFP-LC3 (induction with more than 100 MOI of adenovirus), a strongincrease of the green fluorescence signal could be observed regardlessof whether an activator or an inhibitor of autophagywas applied. There-fore, it is important to achieve a moderate and equal expression of theLC3-conjugated florescence proteins in cellular models cells used forthe assessment of the autophagic flux.

Our time-lapse, live cell imaging further support our observations onsalinomycin-triggered autophagy in the context of an active lysosomalfunction. Salinomycin causes severe vacuolization in the cytoplasm, asobserved in our time-lapse video (Supplementary Fig. 4). The initialmembrane for autophagic vessel formation also originated from sourcessuch as Golgi, endoplasmic reticulum, etc. [13]. Thus, to study autopha-gic flux, we preferred to use GFP-LC3 andmTandemGFP-RFP LC3 ratherthan using Cyto-ID, which depends on membrane composition of thevacuoles [14]. Some studies demonstrate that vacuolization acts as animpediment for the completion of autophagic processes through dis-ruption of lysosomal fusion with autophagosomes [15]. However,since both our studies show no such disruption in the fusion of lyso-somes with the autophagosomes upon salinomycin treatment, andmoreover, that the ability of ionophores to induce vacuolization is celltype dependent, one cannot exclude that excessive vacuolization athigher concentrations of salinomycin could play a secondary role inthe inhibition of autophagic flux [14].

In conclusion, although autophagy is activated as a component of aninitial stress response (cell copingmechanism), under the conditions ofextreme stress that may cause cell death, autophagy could fail to pro-vide protection, and also act as an enhancer of apoptosis. Several ofthe otherwise autophagy-promoting factors like Ambra1, Atg4d andATG5, are differentially regulated upon cell death trigger [16–18].Ambra1, which is mainly responsible for the initiation of autophagy, isdownregulated once the cell is committed to cell death, thus inhibitingautophagy [16]. On the other hand, Atg4d during cell death is localized

Please cite this article as: J.R. Jangamreddy, et al., Monitoring of autophag(2014), http://dx.doi.org/10.1016/j.bbamcr.2014.12.022

ED P

RO

OF

to the mitochondrial membrane while ATG5 nuclear localization triggersmitotic catastrophe [17,18]. Similarly, 10 μM salinomycin treatment ofMCF7 cells for 16 h downregulates Beclin1 and ATG12, which are in-volved in the initiation andelongationof autophagy,while showing an in-crease at initial timepoints (2–8h) and lower doses [19]. This observationfurther supports the dual nature of autophagic response to salinomycintreatment. Taken together, our observations suggest that the inhibitionof autophagic flux by salinomycin is a secondary phenomenon.

4. Materials and Methods

4.1. Cells and Cell culture

PC3 (human prostate cancer cell line), SKBR3 (human breast cancercell lines) and murine embryonic fibroblast (MEF) cells were culturedin complete media (RPMI or DMEM media supplemented with 10%FBS and 1% penicillin-streptomycin antibiotics) and maintained atsubconfluent conditions as mentioned in our previous publication [6].

4.2. Materials and reagents

Salinomycin, rapamycin, rhloroquine and saponin, were obtainedfrom Sigma-Aldrich and dissolved in their respective buffers as perrequired concentrations. Rabbit-anti-LC3b, murine anti-actin was alsofrom Sigma-Aldrich whereas rabbit-anti-p62/SQSTRM1 was obtainedfrom Cellular Signalling Inc. Secondary antibodies such as anti-rabbitHRP-conjugate and anti-murine HRP-conjugates were obtained fromSigma-Aldrich. Adenovirus expressing GFP-LC3 and mTandem GFP-RFP-LC3 were a kind gift from Dr. Junichi Sadoshima (Cell Biologyand Molecular Medicine Rutgers University, New Jersey MedicalSchool, USA).

4.3. Western blot

Cells cultured and treated as per experimental needs were lysedusing 100 μl of lysis buffer (RIPA buffer with protease inhibitors (Com-plete Roche)) and mechanically sheared using 27 G ¾ inch needle andsyringe. The lysate was then centrifuged at 10,000 RPM to pellet thecell debris and supernatant was collected. Protein concentration wasdetermined using Bradford assay and equal amounts of protein sampleswere loaded onto the 15% PAGE gel and further proceeded as describedpreviously [6]. Themembranewas developed usingAmershamECL plusWestern blot developing kit (GE technologies).

4.4. Live cell imaging and confocal microscopy

Cells plated were infected with adenovirus expressing GFP-LC3 at aviral titer of 50 MOI for 24 h and treated with lysotracker (100 nM)for 30 min were treated with various concentrations of salinomycin(0.1 μM to 1 μM) as mentioned accordingly and images were acquiredusing Leica DMI6000 Florescence microscope equipped with a DFC365monochrome camera and 63× oil objective (NA 0.6–1.4) for 1 h at arate of 10 s interval per frame and 8 z-stack slices of the cell is takenafter setting the lower and upper limit of the Z section defining thecell size. The images were further analyzed using ImageJ and BitplaneImaris software. For confocal imaging, cells pre-incubated with adeno-virus expressing GFP-LC3 or mTandem GFP-RFP-LC3 at 50 MOI andtreated as mentioned above were washed with PBS and fixed using 4%paraformaldehyde for 20 min at 4 °C and mounted onto a slide withmounting medium containing DAPI. Images were taken using laserscanning confocal microscope (Zeiss).

4.5. Autophagic flux analysis using flow cytometry

A total of 100,000 cells/well, plated in 6-well plates were culturedfor 24 h and infected with GFP-LC3 or mTandem-GFP-RFP-LC3 for 24

y is complicated—salinomycin as an example, Biochim. Biophys. Acta

T

320

321

322

323

324

325

326

327

328

329

330

331

332

333

334

335

336

337

338

339

340

341

342

343

344

345

346

347348

349350351352353354355356357358359360361362363364365366367368369370371372373374375376377378379380381382383384385386387388389390391392393394395396397

7J.R. Jangamreddy et al. / Biochimica et Biophysica Acta xxx (2014) xxx–xxx

C

or 48 h. Cells were then treated as described in the Results section,to monitor autophagy and autophagic flux, for 6 h. The cells werethen washed with PBS, trypsinized and collected into an eppendorftube. Then, the cells were centrifuged and single cell suspensions weremade with 500 μl of PBS before being analyzed using Gallios FlowCytometer (GFP with FL1 and RFP with FL4 filters both excited using488 laser). FlowJo software was used for analysis and representationof histograms. The GFP-LC3 expressing cells are optionally incubatedwith 0.05% of saponin for 10 min for the respective studies.

4.6. Statistics

All statistics (one-way ANOVA) were conducted using Prism (ver-sion 6.0b) and SPSS (IBM version 20) software. A p-value of less than0.05 was considered statistically significant unless mentioned other-wise. All the experiments were conducted as a minimum of three inde-pendent replicates unless otherwise mentioned.

Acknowledgements

We kindly thank Dr. Thommie Karlsson (application specialist atLeica Microsystems) and Dr. Venkata Ramana Rao Parasa for helpingin the analysis of live cell imaging and using Bitplane Imaris software,respectively. MJL kindly acknowledges the core/startup support fromLinkoping University, from Integrative Regenerative Medicine Center(IGEN), from Cancerfonden (2013/391), and from VR-NanoVision(K2012-99X-22325-01-5).

Appendix A. Supplementary data

Supplementary data to this article can be found online at http://dx.doi.org/10.1016/j.bbamcr.2014.12.022.

References

[1] D. Hanahan, R.A. Weinberg, Hallmarks of cancer: the next generation, Cell 144(2011) 646–674.

UNCO

RRE

Please cite this article as: J.R. Jangamreddy, et al., Monitoring of autophag(2014), http://dx.doi.org/10.1016/j.bbamcr.2014.12.022

ED P

RO

OF

[2] D.J. Klionsky, F.C. Abdalla, H. Abeliovich, R.T. Abraham, A. Acevedo-Arozena, et al.,Guidelines for the use and interpretation of assays for monitoring autophagy,Autophagy 8 (2012) 445–544.

[3] D. Glick, S. Barth, K.F. Macleod, Autophagy: cellular and molecular mechanisms,J. Pathol. 221 (2010) 3–12.

[4] C. Zhou, W. Zhong, J. Zhou, F. Sheng, Z. Fang, Y. Wei, Y. Chen, X. Deng, B. Xia, J. Lin,Monitoring autophagic flux by an improved tandem fluorescent-tagged LC3(mTagRFP-mWasabi-LC3) reveals that high-dose rapamycin impairs autophagicflux in cancer cells, Autophagy 8 (2012) 1215–1226.

[5] P.B. Gupta, T.T. Onder, G. Jiang, K. Tao, C. Kuperwasser, R.A. Weinberg, E.S. Lander,Identification of selective inhibitors of cancer stem cells by high-throughputscreening, Cell 138 (2009) 645–659.

[6] J.R. Jangamreddy, S. Ghavami, J. Grabarek, G. Kratz, E. Wiechec, B.A. Fredriksson, R.K.Rao Pariti, A. Cieslar-Pobuda, S. Panigrahi, M.J. Los, Salinomycin induces activationof autophagy, mitophagy and affects mitochondrial polarity: differences betweenprimary and cancer cells, Biochim. Biophys. Acta 1833 (2013) 2057–2069.

[7] J.R. Jangamreddy, M.J. Los, Mitoptosis, a novel mitochondrial death mechanismleading predominantly to activation of autophagy, Hepat. Mon. 12 (2012) e6159.

[8] T. Li, L. Su, N. Zhong, X. Hao, D. Zhong, S. Singhal, X. Liu, Salinomycin inducescell death with autophagy through activation of endoplasmic reticulum stress inhuman cancer cells, Autophagy 9 (2013) 1057–1068.

[9] W. Yue, A. Hamai, G. Tonelli, C. Bauvy, V. Nicolas, H. Tharinger, P. Codogno, M.Mehrpour, Inhibition of the autophagic flux by salinomycin in breast cancerstem-like/progenitor cells interferes with their maintenance, Autophagy 9 (2013)714–729.

[10] E. Shvets, E. Fass, Z. Elazar, Utilizing flow cytometry to monitor autophagy in livingmammalian cells, Autophagy 4 (2008) 621–628.

[11] P. Hundeshagen, A. Hamacher-Brady, R. Eils, N.R. Brady, Concurrent detectionof autolysosome formation and lysosomal degradation by flow cytometry in ahigh-content screen for inducers of autophagy, BMC Biol. 9 (2011) 38.

[12] K.E. Eng, M.D. Panas, G.B. Karlsson Hedestam, G.M. McInerney, A novel quantitativeflow cytometry-based assay for autophagy, Autophagy 6 (2010) 634–641.

[13] S.A. Tooze, T. Yoshimori, The origin of the autophagosomalmembrane, Nat. Cell Biol.12 (2010) 831–835.

[14] G. Morissette, E. Moreau, C.G. R, F. Marceau, Massive cell vacuolization induced byorganic amines such as procainamide, J. Pharmacol. Exp. Ther. 310 (2004) 395–406.

[15] C.L. Oeste, E. Seco, W.F. Patton, P. Boya, D. Perez-Sala, Interactions between autopha-gic and endo-lysosomal markers in endothelial cells, Histochem. Cell Biol. 139(2013) 659–670.

[16] G.M. Fimia, M. Corazzari, M. Antonioli, M. Piacentini, Ambra1 at the crossroadbetween autophagy and cell death, Oncogene 32 (2013) 3311–3318.

[17] V.M. Betin, J.D. Lane, Atg4D at the interface between autophagy and apoptosis,Autophagy 5 (2009) 1057–1059.

[18] D. Maskey, S. Yousefi, I. Schmid, I. Zlobec, A. Perren, R. Friis, H.U. Simon, ATG5 isinduced by DNA-damaging agents and promotes mitotic catastrophe independentof autophagy, Nat. Commun. 4 (2013) 2130.

[19] B. Verdoodt, M. Vogt, I. Schmitz, S.T. Liffers, A. Tannapfel, A. Mirmohammadsadegh,Salinomycin induces autophagy in colon and breast cancer cells with concomitantgeneration of reactive oxygen species, PLoS ONE 7 (2012) e44132.

y is complicated—salinomycin as an example, Biochim. Biophys. Acta